Metabolic Effects of Psychotropic Drugs
Modern Trends in Pharmacopsychiatry Vol. 26
Series Editor
B.E. Leonard
Galway
Metabolic Effects of Psychotropic Drugs Volume Editors
J. Thakore Dublin B.E. Leonard Galway 6 figures and 11 tables, 2009
Basel · Freiburg · Paris · London · New York · Bangalore · Bangkok · Shanghai · Singapore · Tokyo · Sydney
Modern Trends in Pharmacopsychiatry Formerly published as ‘Modern Problems in Pharmacopsychiatry’
Dr. Jogin Thakore
Prof. Brian E. Leonard
Neuroscience Centre St. Vincent’s Hospital Fairview Dublin (Ireland)
Department of Pharmacology National University of Ireland Galway (Ireland)
Library of Congress Cataloging-in-Publication Data Metabolic effects of psychotropic drugs / volume editors, J. Thakore, B.E. Leonard. p. ; cm. – (Modern trends in pharmacopsychiatry, ISSN 1662-2685 ; v. 26) Includes bibliographical references and index. ISBN 978-3-8055-9001-3 (hard cover : alk. paper) 1. Psychotropic drugs–Side effects. 2. Metabolic syndrome. 3. Schizophrenia–Chemotherapy–Complications. I. Thakore, Jogin H., 1962– II. Leonard, B. E. III. Series: Modern trends in pharmacopsychiatry ; v. 26. [DNLM: 1. Psychotropic Drugs–adverse effects. 2. Psychotropic Drugs–metabolism. 3. Mental Disorders–drug therapy. W1 MO168P v.26 2009 / QV 77.2 M587 2009] RM315.M456 2009 615'.788–dc22
Bibliographic Indices. This publication is listed in bibliographic services, including Current Contents® and Index Medicus. Disclaimer. The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publisher and the editor(s). The appearance of advertisements in the book is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements. Drug Dosage. The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any change in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug. All rights reserved. No part of this publication may be translated into other languages, reproduced or utilized in any form or by any means electronic or mechanical, including photocopying, recording, microcopying, or by any information storage and retrieval system, without permission in writing from the publisher. © Copyright 2009 by S. Karger AG, P.O. Box, CH–4009 Basel (Switzerland) www.karger.com Printed in Switzerland on acid-free and non-aging paper (ISO 9706) by Reinhardt Druck, Basel ISSN 1662-2685 ISBN 978–3–8055–9001-3
Contents
VII Preface 1 12 25 47
66
82
90
105 116
129 130
Metabolic Syndrome and Schizophrenia: Is Inflammation the Cause? Leonard, B.E. (Galway/Munich) Insulin Resistance in Bipolar Women: Effects of Mood-Stabilizing Drugs Vemuri, M.; Stemmle, P.; Jiang, B.; Morenkova, A.; Rasgon, N. (Stanford, Calif.) Obesity and Mental Illness Citrome, L. (Orangeburg, N.Y.); Vreeland, B. (Piscataway, N.J.) Glucose Abnormalities in Schizophrenia, Bipolar and Major Depressive Disorders Bushe, C. (Basingstoke) Screening for Diabetes and Cardiovascular Risk Factors in People with Serious Mental Illness Holt, R.I.G.; Peveler, R.C. (Southampton) Stress Axis Dysfunction: A Common Finding in Schizophrenia and the Metabolic Syndrome? Afzal, A.; Thakore, J. (Dublin) Metabolic Syndrome in Psychiatric Inpatients Treated for Depression Goethe, J.W.; Szarek, B.L. (Hartford, Conn.); Caley, C.F. (Hartford, Conn./Storrs, Conn.) Hyperprolactinaemia Associated with Antipsychotic Medications Fitzgerald, P.; Dinan, T.G. (Cork) Impact of Hyperprolactinaemia on the General Health of Patients with Schizophrenia Wildgust, H.J. (Ackworth).; Kohen, D. (Preston) Author Index Subject Index
V
Preface
‘There are no psychiatric patients, only medical patients with varying degrees of psychopathology.’ Schiffer RB, Klein RF, Sider RC: In: The Medical Evaluation of Psychiatric Patients. New York, Plenum, 1988, p. 28. Patients suffering from major psychiatric disorders have reduced life expectancy. It is now well established that the increased morbidity and mortality is often caused by cardiovascular disease, cancer, inflammatory diseases and endocrine disorders such as type 2 diabetes. Yet it is only in recent years that attempts have been made to integrate the causes of physical illness with the underlying psychopathology of the disorder. Thus, it is now recognized that dysfunctional metabolism plays a crucial role in physical ill health that is associated with chronic psychiatric disorders such as depression and schizophrenia. Whether the metabolic syndrome is primarily caused by changes resulting from life style or the pathophysiology of the patient or, alternatively, is an important contribution to the psychopathology of the psychiatric disorder is an open question. A further complication relates to the impact of the drugs on the metabolism. Nowhere is this more apparent than in the reported effects of some atypical antipsychotics and antidepressants in causing weight gain, and possibly type 2 diabetes. Yet, as is apparent from several chapters in this volume, it is uncertain whether the drugs cause the metabolic syndrome or that they exacerbate a vulnerability that exists in some major psychiatric disorders. In this volume, the editors have brought together experts from clinical psychiatry, epidemiology, neuroimmunology, endocrinology and neuroscience to explore the different facets of the metabolic syndrome and its connection with the chronic effects of psychotropic drugs and with the psychopathology of some major psychiatric disorders.
VII
Leonard puts forward the hypothesis that schizophrenia may be a low grade chronic inflammatory disorder by weaving together its possible viral, neurodevelopmental origins with recent evidence showing changes in various inflammtory markers such as IL-6 and TNF. Vemuri and coworkers focus on the important topic of insulin resistance and bipolar disorder with obesity, lifestyle choices and mood stabilizers playing important roles in women suffering from this mood disorder. Citrome and Vreeland discuss the topical and at times controversial issue of why obesity may develop in those mental illness and offer pertinent and practical solutions in the form of ‘small steps’ that may help the vast majority of patients to gain control over their weight and also discuss the possibility of using pharmacological and surgical interventions while Bushe focuses on the glucose abnormalities observed within chronic psychiatric disorders and the role of the illness versus antipsychotic agents. The author of this chapter poignantly observes that we still have not unraveled this complex relationship despite the explosion of information that we have witnessed over the last years. Based upon criteria set forth by the UK National Screening Committee, Holt and Peveler offer a compelling set of reasons for why type 2 diabetes and cardiovascular disease should be screened for in those with mental illness. Afzal and Thakore explore the relationship between stress, schizophrenia and the metabolic syndrome showing that a dysfunciotnal hypothalamic-pituitary-adrenal axis may be a common occurrence in both conditions and be partly responsible for their respective psychopathological and physical manifestations. The chapter by Goethe and coworkers offers important metabolic insights into probably the most common psychiatric illness, namely depression. This chapter is a study which in effect details the rates of the metabolic syndrome in major depression and the various associations that may be related. In particular they find that atypical antipsychotics may not be associated with the higher than expected rates observed. Fitzgerald and Dinan then focus on how certain antipsychotics decrease dopaminergic activity leading to an increase in prolactin release which has potentially serious adverse effects ranging from lowering bone mineral density which can lead to fractures to being associated with breast and prostate cancer. Wildgust and Kohen discuss the propensity of antipsychotics to induce hyperprolactinemia which in turn can induce a lowering of bone mineral density, gynecomastia, galactorrhea and various menstrual cycle changes resulting in adverse hormonal profiles that could possibly potentiate the inherent risk that those with psychiatric illnesses have to cardiovascular disease. This is the first monograph in a series devoted to pharmacopsychiatry. The aim of the series is to consider the inter-relationship between psychotropic drugs and the underlying psychopathology of psychiatric disorders. The current volume will consider the various aspects of the metabolic syndrome in major psychiatric disorders. Hopefully the contents of this first volume will be of interest not only to clinical psychiatrists but also to endocrinologists, immunologists, cardiologists and clinical neuroscientists. Jogin Thakore Brian E. Leonard
VIII
Preface
Thakore J, Leonard BE (eds): Metabolic Effects of Psychotropic Drugs. Mod Trends Pharmacopsychiatry. Basel, Karger, 2009, vol 26, pp 1–11
Metabolic Syndrome and Schizophrenia: Is Inflammation the Cause? Brian E. Leonarda,b a
Pharmacology Department, National University of Ireland, Galway, Ireland; bDepartment of Psychiatry and Psychotherapy, Ludwig Maximilians University, Munich, Germany
Abstract Physical ill health is a common feature of schizophrenia. Hypertension, elevated cholesterol, obesity and type 2 diabetes are common occurrences and lead to increased morbidity and mortality. The question therefore arises whether these changes are due to an unhealthy life style of the patient or to a genetic predisposition that is exacerbated by life style and/or psychotropic medication. However, it is also possible that the metabolic changes that underlie physical ill health are a reflection of immune dysfunction. This review considers the evidence implicating an increase in inflammation in the psychopathology of schizophrenia, this being major contributory cause of diabetes and cardiovascular disease. It is concluded that there is substantial evidence to support this hypothesis and that more research is needed into the causal links between metabolic dysfunction and the psychopathology of schizophrenia in order to improve the physical, as well as the mental health, of the patient. Copyright © 2009 S. Karger AG, Basel
It is generally recognized that the ill health of the schizophrenic patient consists not only of psychiatric symptoms but also of physical illness. Thus, it has been estimated that the life expectancy for patients with schizophrenia is 20% lower that of the general population. Such premature death can result from increased risk of heart and cerebrovascular diseases while psychiatric co-morbidity includes alcohol and drug abuse which also contribute to the general ill health of the patient [1, 2]. Schizophrenia is associated with hypertension, obesity and elevated cholesterol [3]. Not surprisingly, the rates of the metabolic syndrome, type 2 diabetes and cardiovascular disease are higher than expected. The question therefore arises whether the increase in such conditions is a reflection of the patients unhealthy life style (lack of exercise and poor diet for example) or due to a genetic predisposition that is exacerbated by certain types of psychotropic medication. Furthermore, the increase in blood pressure, elevated cholesterol and diabetes are commonly associated with obesity in schizophrenic patients [3]. However, are these connections causal or co-incidental? If they are causally related,
are they primarily due to the type of antipsychotic drug being administered to the patient and, if so, are some antipsychotics more likely to cause the metabolic syndrome than others? If the metabolic and physical changes are co-incidental then, presumably, by changing the life style of the patient such adverse changes should be reduced. These are some of the questions raised by the authors in this volume. It is possible that the metabolic changes associated with schizophrenia are a reflection of immune dysfunction. Only in recent years has research established that there are clinically significant changes in the aspects of cellular and humoral immunity associated with the major symptoms of psychiatric disorders such as schizophrenia, bipolar disorder and depression [4, 5]. Furthermore, it is apparent that effective treatment of these disorders with psychotropic drugs largely attenuates the aberrant changes. The purpose of this chapter is to consider the possible involvement of inflammation as a common, possibly causal factor, in both the physical disorders and clinical symptoms that can accompany schizophrenia (fig. 1)
Immune Changes in Schizophrenia
The concept that a dysfunctional immune system plays a role in the etiology of schizophrenia can be traced back to the 19th century when it was estimated that approximately one-third of psychotic patients in Europe were suffering from neurosyphilis, a condition that has largely disappeared following the introduction of antibiotics in the 20th century. However, in the last century, it was observed that viral infections such as rubella and the influenza virus were also associated with the symptoms of schizophrenia, at least in some patients [6]. Perhaps the most compelling evidence for a link between schizophrenia and a dysfunctional immune system has been provided by Lindholm et al. [7] who demonstrated that a locus at chromosome 6p22 was linked to both schizophrenia and to the genes of the human lymphocyte (HLA) system. This system is crucially involved in combating viruses which might account for the increased vulnerability of schizophrenic patients to viral infections. Another interesting connection between schizophrenia and the immune system has been provided by the susceptibility of first-degree relatives of schizophrenic patients to insulin-dependent diabetes mellitus. This susceptibility appears to be associated with the HLA locus on chromosome 6 [8]. Such an observation is particularly important because of the association between the metabolic syndrome and type 2 diabetes in schizophrenic patients being treated with the second-generation antipsychotic olanzapine. Clearly, there are differences in the genetic signal between type 1 and type 2 diabetes. Thus, there appears to be a decrease in the frequency of schizophrenia in patients with type 1 diabetes [9], which suggests that insulin, as such, is not directly linked to the pathology of schizophrenia. As is becoming apparent, type 2 diabetes is a possible consequence of dysfunctional metabolism initiated by immune, endocrine and mitochondrial changes that are symptomatic of schizophrenia.
2
Leonard
Nutrient excess
Energy input ⬍⬍ energy output
Oxidative stress (long-chain fatty acid accumulation; fat depots)
Obesity Inflammation
Insulin resistance
Metabolic disorders (cardiovascular disease; type 2 diabetes, etc.) Fig. 1. Link between inflammation, obesity, diabetes and cardiovascular disease in schizophrenic patients.
Despite the circumstantial evidence linking viral infections to schizophrenia, no single virus has so far been identified as a causal agent. Furthermore, there is no evidence of a major immune reaction in the brains of schizophrenic patients; for example, no marked gliosis or evidence of lymphocyte infiltrates even though there is evidence of diffuse humoral reactivity in some patients. In addition, there is evidence that some of the peripheral immune changes are reflected in the brain [10, 11]. Leonard [10] has critically reviewed the evidence linking a disorder of the immune system to the psychopathology of schizophrenia
Evidence for the Activation of Cellular Immunity in Schizophrenia
In schizophrenia, there is evidence that the innate immune system is activated. Thus, the number of monocytes and some of the cytotoxic cells are increased [12]. In addition, the proportion of monocytes and macrophages in the CSF of patients with acute schizophrenia are increased, suggesting that immune activation occurs in the brain as well as in the periphery [13]. Of the pro-inflammatory cytokines that are raised in the CSF, interleukin 6 (IL-6) has been reported to be increased by a number of investigators [14]. IL-6 activates B cells, in addition to playing a key role in the inflammatory cytokine cascade, and therefore contributes to many of the immune changes seen in schizophrenia. Il-6, in addition to other pro-inflammatory cytokines, is released from activated macrophages and monocytes in the periphery and from microglia and astrocytes in
Metabolic Syndrome and Schizophrenia
3
the brain. In vitro evidence has shown that IL-6 also stimulates the release of prolactin from the pituitary [15]. Whether this is relevant to the increase in the secretion of prolactin in patients being treated with antipsychotics is questionable as effective antipsychotic treatment usually attenuates the secretion of pro-inflammatory cytokines. If IL-6 plays a role in the psychopathology of schizophrenia, to what extent do changes in this cytokine reflect the clinical status of the patient? It is clear that IL-6 is raised in the plasma of schizophrenic patients [15, 16] and that elevated plasma concentrations of this cytokine are related to both the duration of the disorder [17] and resistance to drug treatment [18]. In the CSF, the concentration of the soluble IL-6 receptor (sIL-6R) is also increased [18]. Il-6 has also been shown to activate both dopaminergic and serotonergic neurons in the hippocampus and the frontal cortex [19]. As these neurotransmitter changes have been implicated in the etiology of schizophrenia, it would appear that there is a close inter-relationship between the activation of the central and peripheral immune system and the changes in the monoamine neurotransmitters that are thought to be involved in the pathology of the disorder. Although marked gliosis has seldom been detected in schizophrenic patients, microglial activation, triggered by the increase in the pro-inflammatory cytokines IL-1 and IL-6, has been detected in the frontal cortex [20]. It has also been observed that the expression of the IL-1 receptor antagonist (IL-1RA) is decreased in the prefrontal cortex of schizophrenic patients; this antagonist counteracts the overstimulation of IL-1 receptors by IL-1 [21]. As a consequence of the overactivation of IL-1, the hypothalamic-pituitary-adrenal axis is also activated; IL-1 is known to activate the anterior pituitary thereby enhancing the stress response [22]. IL-1 also decreases long-term potentiation in the hippocampus [23] thereby contributing to the disordered memory function frequently seen in patients with schizophrenia. In addition to IL-6 and IL-1, other cytokines also appear to change in the plasma of schizophrenic patients. The changes in IL-2 and interferon-γ (IFN) were reported to be decreased, at least in some large-scale studies [24]. These findings suggest that there is a decrease in the Th-1 arm of the cellular immune system in patients with schizophrenia. It must be emphasized though that there are several conflicting findings in the literature regarding this conclusion [4, 25]. In the CSF, there is evidence that the IL-2 concentrations are increased in those patients who relapsed following treatment with haloperidol; these changes were associated with the recurrence of psychotic symptoms [24]. This may suggest that there is not a direct relationship between the blood and brain compartments of the immune system regarding the role of IL-2. On balance, it would appear that some schizophrenic patients show a shift in the adaptive immune system from cellular (Th1-mediated) to humoral (Th2-mediated) immunity. This shift appears to be more prominent in patients with predominantly negative symptoms that show a poor response to antipsychotic therapy [4, 25]. Schizophrenia is frequently considered to be a neurodevelopmental disorder that may arise as a consequence of prenatal exposure to a virus. Microglia are known to
4
Leonard
migrate into the brain early in development and to be involved in neural growth (low concentrations of pro-inflammatory cytokine have neuronal growth factor potential), pruning of neurons and removal of cell debris. Microglia therefore fulfil the role of macrophages in the brain and are involved in the presentation of antigens to phagocytic cells. They also release pro-inflammatory cytokines and are known to play a crucial role in immune function in schizophrenia so that an overactivation of the microglia contributes to the increased inflammatory challenge to the brain. Conversely, the astroglial cells have a largely neuroprotective role in the brain. There is evidence that the concentration of the S100 beta protein, derived from astroglial cells, is raised in patients independently of their medication [26], which has led to the suggestion that this change is in response to the neurodegenerative impact of the inflammatory mediators produced by the activated microglia.
Effect of Antipsychotic Drugs on the Immune Response in Schizophrenic Patients
Both in vitro and in vivo studies have suggested that effective antipsychotic treatment corrects the imbalance between the cellular and humoral arms of the adaptive immune system [4, 27]. Both haloperidol and clozapine, for example, have been shown to increase the release of IL-2 and IFN from whole blood cultures in vitro, whereas the antidepressant amitriptyline was ineffective [28]. In vivo, IL-18, a cytokine that also originates from activated Th-1 cells, is also increased following effective antipsychotic treatment, while the blunted reaction of patients to a salmonella vaccine challenge is reversed following effective drug treatment. In addition, the Th-3 cells have recently been implicated in correcting the imbalance between the Th-1 and Th-2 arms of the adaptive immune system. The Th-3 cells secrete transforming growth factors (TGF) of which serum TGF-1β has been reported to be elevated in schizophrenia [29]. This suggests that antipsychotic drug-induced changes in the concentration of TGF-1β might play a role in the normalization of the imbalance between the Th-1 and Th-2 arms of the immune system. The activation of the B cells in schizophrenic patients to produce antibodies is an important part of humoral immunity and it would be anticipated that effective drug treatment would be correlated with an increase in antibody titer. There is ample clinical evidence that the antibody response to a vaccine challenge is reduced in schizophrenic patients but normalized on effective treatment [30]. Extensive studies have demonstrated that both the CD5 and B cells increase during long-term treatment with antipsychotic drugs [31]. Such findings lend further support to the view that there is an imbalance between the Th-1 and Th-2 systems which is corrected by effective antipsychotic treatment. If inflammation plays a crucial role in the psychopathology of schizophrenia, then it would seem reasonable to postulate that anti-inflammatory drugs should have a therapeutic effect on the symptoms of the disorder. Prostaglandin E2 is a major
Metabolic Syndrome and Schizophrenia
5
inflammatory mediator in the brain and is synthesized by cyclo-oxygenase (COX) from arachidonic acid. Of the two COX enzymes in the brain, COX-2 is induced by pro-inflammatory cytokines. The product, prostaglandin 2, enhances the synthesis of Th-2 cytokines and inhibits the synthesis of Th-1 cytokines [32]. Of the COX-2 inhibitors that are available for clinical use in the treatment of arthritis, celecoxib is the most lipophilic. Recent clinical studies have demonstrated that in a prospective, double-blind, randomized trial of risperidone, alone or in combination with celecoxib, celecoxib significantly improved the positive and negative symptoms of the schizophrenic patients relative to those treated with risperidone alone [33]. This initial observation will hopefully lead to further studies of the use of anti-inflammatory agents in the treatment of schizophrenia.
On the Possible Role of Inflammation in Obesity and Type -2 Diabetes in Schizophrenia
It is well known that genetic and environmental factors interact to favor weight gain, changes which disrupt metabolism. The body fat stores are normally maintained within a narrow range by energy homeostasis. This process is controlled by the brain regions, such as hypothalamus, that control appetite and energy balance in addition to peripheral signaling systems that monitor energy stores. Glucose, free fatty acids, insulin and leptin are examples of the signaling molecules that activate the hypothalamus thereby controlling the metabolic rate and the desire to eat. Obesity does not simply arise from the passive accumulation of excess body weight but is an active adaptation to the elevation of body fat. Clearly, the genetic background of the individual contributes to the variation in the response to elevated body fat which helps to explain why some individuals are protected against weight gain while the majority is not despite the fact that they live in the same environment and eat the same food. When energy intake exceeds expenditure, the resultant state of nutrient excess triggers responses in vascular endothelial cells, hepatocytes, myocytes, adipocytes, monocytes and macrophages resulting in metabolic dysfunction [34]. The cellular responses to the nutrient excess include the production of reactive oxygen species (ROS) that are generated by the mitochondrial oxidation of glucose and fatty acids. The resulting oxidative stress frequently results in cellular damage and triggers the inflammatory response [35]. Long-chain fatty acids and co-enzyme Q derivatives that are usually metabolized by the mitochondria also accumulate under these conditions and reflect a decrease in mitochondrial oxidative function. The final result of these changes is an activation of the C-jun N-terminal kinase and kappa-B pathways that promote inflammation. Thus inflammation appears to be a common end point of obesity. The link between obesity and diabetes occurs as a result of a reduction in insulin function, thereby reducing the uptake of glucose into the target tissues. It is established that phosphotidylinositol-3-hydroxykinase, the inositol receptor substrate pathway, is
6
Leonard
particularly sensitive to inactivation by inflammatory mediators [36, 37]. Thus, with continuing nutrient excess tissues become insulin insensitive thereby leading to an extension of insulin resistance to most tissues as the inflammatory state progresses. In addition to regulating nutrient utilization in peripheral tissues, the insulin receptor signaling system has been implicated in the actions of insulin and lepin on neuronal function. From experimental studies, it would appear that even short-term exposure to a palatable, energy-rich diet impairs the responsiveness of the hypothalamus to the effects of insulin and leptin [38]. This reduces the feedback regulatory mechanism whereby the brain normally reduces food intake in response to the increase in peripheral fat stores. From the foregoing, it can be seen how inflammation participates in the link between obesity and type-2 diabetes. The ‘beta-cell exhaustion hypothesis’ of diabetes postulates that diabetes results when pancreatic beta cells fail to produce sufficient insulin due to insulin resistance thereby initiating type-2 diabetes [39]. Obesity is also linked to cardiovascular disease, both conditions occurring frequently in patients with schizophrenia. This is due to the impact of nutrient excess on the vascular endothelial cells. A major function of these cells is to activate nitric oxide synthase to produce the potent, but short-lived, vasodilator nitric oxide. The insulin receptor system-phosphoinositol-3-kinase pathway is a major factor in the control of nitric oxide synthesis in most tissues so that nutrient excess that reduces the activity of this pathway rapidly leads to a reduction in nitric oxide synthesis. Thus, the decrease in nitric oxide, combined with the accumulation of fat in the coronary circulation as a consequence of increased fat synthesis, provides a link between cardiovascular disease, diabetes obesity and inflammation [40]. While it is unlikely that the inhibition of the insulin-sensitive pathway by inflammatory mediators is the only factor of importance, it does emphasize the value of the integrative approach to studying metabolic disease and how such an approach may contribute to the physical ill health associated with major psychiatric disorders such as schizophrenia.
Role of Leptin in Inflammation and Its Possible Connection to Obesity in Schizophrenia
In most obese patients, obesity is associated with chronic low-grade inflammation of white adipose tissue. This results from the chronic activation of the innate immune system that subsequently leads to insulin resistance, impaired glucose tolerance and diabetes [41]. In obesity, the white adipose tissue is characterized by the increased synthesis and secretion of a range of inflammatory mediators such as tumor necrosis factor (TNF) and IL-6. In addition, the adipose tissue is infiltrated by macrophages that act as a major source of pro-inflammatory cytokines when activated. Conversely, weight loss leads to a reduction in the gene expression of the pro-inflammatory
Metabolic Syndrome and Schizophrenia
7
cytokines [42]. However, the adipocytes and macrophages are responsible for the increase not only of the pro-inflammatory cytokines but also for leptin and resistin. The increase in leptin exerts a strong negative feedback on insulin sensitivity thereby enhancing insulin resistance [43]. This situation is compounded by the impact of the glucocorticoids on the regulation of leptin; hypercortisolemia, a common feature of schizophrenia and other major psychiatric disorders, stimulates the release of leptin and thereby further enhances insulin resistance [44]. As has been mentioned previously, premature death due to cardiovascular disease is a common occurrence in both schizophrenia and depression. IL-6 is increased in both schizophrenia and major depression and is known to induce hepatic C-reactive protein that has been implicated in cardiovascular disease [45]. In addition to such changes, obesity is associated with a decrease in circulating adiponectin. Adiponectin is a cardioprotective agent that reduces the inflammatory action of pro-inflammatory cytokines such as TNF on the arterial walls and thereby protects against atherosclerosis [46].Thus a reduction in the availability of adiponectin in obesity indirectly contributes to an increase in vulnerability to cardiovascular disease. Figure 2 summarizes the link between impaired immunity, cardiovascular disease and obesity in schizophrenic patients. Irrespective of the cause, epidemiological evidence suggests that obesity increases the risk of autoimmune disease and such immune related diseases as asthma. It has been speculated that this arises due to the decrease in immunological tolerance associated with the increase in the pro-inflammatory cytokines and leptin, and the decrease in adiponectin [47]. It has been demonstrated that both IL-6 and leptin downregulate the regulatory T cells thereby reducing antigen surveillance. Thus, obesity, through the induction of chronic low-grade inflammation and decreased immunological tolerance to antigens, increases the activity of the Th-2 humoral pathway thereby increasing the risk of allergies and immune-related disorders. Such changes are a common feature of schizophrenia. Thus, in patients with schizophrenia a vicious cycle is created due to the inflammatory changes. These changes not only impact on the brain to contribute to the psychopathology of the disorder but are also responsible for the decline in physical health that characterizes such patients.
Conclusion
While physical ill health, commonly associated with schizophrenia, is frequently attributed to an unhealthy life style associated with lack of exercise, poor diet and smoking, it is now becoming apparent that there are underlying metabolic abnormalities that predispose patients to obesity and subsequent ill health. Superimposed on the metabolic abnormalities, antipsychotic drugs can contribute to the problem but it is presently unclear whether such drugs are the initiating cause of obesity, type
8
Leonard
(⫹) Inflammation
Schizophrenia
Macrophages, T2 lymphocytes
Obesity
Hypercortisolism
(⫹) White adipose tissue Insulin resistance
(⫹) IL-6 ⫹ TNF-␣ ⫹ IL-10
(⫹) Hepatic CRP
(⫹) Leptin, resistin: (⫺) adiponectin (⫹) T cytokines
Type 2 diabetes
Immune intolerance (⫹) Cardiovascular disease (⫹) Allergies Fig. 2. Link between impaired immunity, obesity, diabetes and cardiovascular disease in patients with schizophrenia. (⫹) ⫽ Increase, (⫺) ⫽ decrease.
2 diabetes and heart disease. Clearly, there is now convincing evidence that there is an underlying disturbance of inflammatory pathways that initiates aberrant metabolic changes. While there is some evidence that effective treatment with antipsychotic drugs may attenuate such changes, it is clinically evident that a substantial proportion of patients still experience physical ill health. Whether the inflammatory changes are attenuated in such patients or not is unknown. Nevertheless, the results of such studies emphasize the need to expand research to investigate the broader aspects of metabolic function with a goal of developing treatments that are not only effective in ameliorating the psychiatric symptoms but also in improving the physical health of the patient.
References 1
2
3
Birkhofer A, Alger P, Schmid G, Foestl H: The cardiovascular risk of schizophrenic patients. Neuro psychiatry 2007;21:261–266. Kozaric-Kovacic D, Folnegovic-Smak V, Folnegovic Z, Maruic A: Influence of alcoholism on the prognosis of schizophrenic patients. J Stud Alcohol 1995; 56:622–627. Birkenaes AB, Birkeland KI, Engh JA, et al: Dyslipidaemia independent of body mass in antipsychotic treated patients under real life conditions. J Clin Psychopharmacol 2008;28:132–137.
Metabolic Syndrome and Schizophrenia
4
5 6
Schwartz MJ, Mueller N, Riedel M, Ackenhail M: Markers of cellular and humoral immune activity: a shift in schizophrenia. Adv Biol Psychiatry 2001;20: 61–65. Smith R: The macrophage theory of depression. Med Hypotheses 1991;35:298–306. Moises HW, Ruger R, Reynolds GP, Fleckenstein B: Human cytomegalovirus DNA in the temporal cortex of a schizophrenic patient. Eur Arch Psychiatry Neurol Sci 1988;238:110–113.
9
7 Lindholm E, Elsholm R, Baleinniene J, et al: Linkage analysis of a large Swedish kindred provides further support for a susceptibility locus for schizophrenia on chromosome 6p23. Am J Med Genet 1999;88: 369–377. 8 Badenhoop K, Tonjes RR, Row H, et al: Endogenous retroviral long-terminal repeats of the HLA-DQ region are associated with susceptibility to insulin dependent diabetes mellitus. Hum Immunol 1996; 50:103–110. 9 Juvonen H, Reunanen A, Haukka J, et al: Incidence of schizophrenia in a nationwide cohort of patients with type 1 diabetes mellitus. Arch Gen Psychiatry 2007;64:894–899. 10 Mueller N, Ackenheil M: Psychoneuroimmunology, the cytokine network in the CNS and implication for psychiatric disorders. Prog Neuropsychopharmacol Biol Psychiatry 1998;22:1–31. 11 Leonard BE: Is there an immunological basis for schizophrenia? Expert Opin Clin Immunol 2005;1: 103–112. 12 Mueller N, Schlesinger BC, Hadjaniu M, et al: Cytotoxic T cells are elevated in unmedicated schizophrenic patients and related to blood-brain barrier and HLA allele DPA 02011. Schizophren Res 1998;12:69–71. 13 Nikkila HV, Muller K, Ahokas A, et al: Accumulation of macrophages in the CSF of schizophrenic patients during acute psychotic episodes. Am J Psychiatry 1999;156:1725–1729. 14 Maes M, Bosman E, CalabreseJ, et al: IL-2 and IL-6 in schizophrenia and mania: effects of neuroleptics and mood stabilisers. J Psychiatr Res 1995;29:141– 152. 15 Spangelobl M, Judd AM, Isakson PC, MacLeod RM: IL-6 stimulates anterior pituitary hormone release in vitro. Endocrinology 1989;125:575–577. 16 Frommberger UH, Bauer J, Haselbauer P, et al: IL-6 plasma levels in depression and schizophrenia: comparison between the acute state and after remission. Eur Arch Psychiatr Clin Neurosci 1997;247: 228–232. 17 Ganguli R, Yang Z, Shurin G, et al: Serum IL-6 concentrations in schizophrenia: elevation associated with duration of illness. Psychiatr Res 1994;51:1–10. 18 Mueller N, Dobmeier P, Empel M, et al: Soluble IL-6 receptor in serum and CSF of paranoid schizophrenic patients. Eur Psychiatry 1997;12:294–299. 19 Zalcman S, Green-Johnson JM, Murray L, et al: Cytokine specific central monoamine alterations induced by IL-1,IL-2 and IL-6. Brain Res 1994;643: 40–49. 20 Radewicz K, Garey LS, Gentleman SM, Reynolds R: Increase in HLA-DR immunoreactive microglia in frontal and temporal cortex in chronic schizophrenics. J Neuropath Exp Neurol 2000;59:137–150.
10
21 Akiyama K: Serum levels of sIL-2R,IL-6 and IL-1RA in schizophrenics before and during neuroleptic administration. Schizophren Res 1999;37:97–106. 22 Sapolsky R, Rivier C, Yamamoto G, et al: IL-1 stimulates the secretion of hypothalamic CRF. Science 1987;238:522–524. 23 Vereker E, O’Donnell E, Lynch MA: The inhibitory effect of IL-1 beta on LTP is coupled with increased activity of stress activated protein kinase. J Neurosci 2000;20:6811–6819. 24 Arolt V, Rothermundt M, Wandinger KP, Kirchner H: Decrease in vitro production of interferon gamma and IL-2 in whole blood of patients with schizophrenia during treatment. Mol Psychiatry 2000;5:150–158. 25 Ganguli R, Brar JS, Chengappa KR, et al: Mitogen stimulated IL-2 production in never medicated first episode schizophrenics: the influence of age of onset and negative symptoms. Arch Gen Psychiatry 1995;52:878–882. 26 Rothermundt M, Ponath G, Glaser T, et al: S100 beta serum levels and long term improvement of negative symptoms in patients with schizophrenia. Neuropsychopharmacology 2004;29:1004–1011. 27 Cazzullo CL, Sacchetti E, Galluzzo A, et al: Cytokine profile in schizophrenic patients treated with risperidone: a three month follow-up study. Prog Neuropsychopharmacol Biol Psychiatry 2002;26:33– 39. 28 Mueller N, Empel M, Riedel M, et al: Neuroleptic treatment increases sIL-2R and decreases IL-6R in schizophrenics. Eur Arch Psychiatr Clin Neurosci 1997;247:308–313. 29 Kim Y-K, Myint A-M, Lee B-H, et al: Th1,Th2 and Th3 cytokine alterations in schizophrenia. Neuropsychopharmacol Biol Psychiatry 2004;28: 1129–1134. 30 Ganguli R, Rabin BS, Kelly RH, et al: Clinical and laboratory evidence of autoimmunity in acute schizophrenia. Ann NY Acad Sci 1987;496:676–685. 31 Mueller N, Riedel M, Schwarz M, et al: Immunomodulatory effects of neuroleptics on the cytokine and cellular immune system in schizophrenia; in Wieselmann G (ed): Current Update in Psychoimmunology. Wien, Springer, 1992, pp 55–62. 32 Hinson RM, Williams JA, Shacter E: Elevated IL-6 induced by PGE2 in a murine model of inflammation: possible role of COX 2. Proc Natl Acad Sci USA 1996;93:4885–4890. 33 Mueller N, Riedel M, Scheppack E, et al: Beneficial antipsychotic effects of celecoxib add-on therapy compared to risperidone alone in schizophrenia. Am J Psychiatry 2002;159:1029–1034. 34 Wisse BE, Kim F, Schwartz MW: An integrative view of obesity. Science 2007;318:928–929.
Leonard
35 De Souza CT, Araujo EP, Bordin S, et al: Consumption of a fat rich diet activates a proinflammatory response and induces insulin resistance in the hypothalamus. Endocrinology 2005;146: 4192–4199 36 Hotamisligil GS: Inflammation and metabolic disorders. Nature 2006;444:860–867. 37 Lowell BB, Shulman GI: Mitochondrial dysfunction and type 2 diabetes. Science 2005;307:384–387. 38 Muenzberg H: Differential leptin access into the brain: a hierarchical organization of hypothalamic leptin target sites? Physiol Behav 2008;94:664–669. 39 Pan DA, Lillioja S, Kriketos AD, et al: Skeletal muscle triglyceride levels are inversely related to insulin action. Diabetes 1997;46:983–988. 40 Kim MJ, Maachi M, Debard C, et al: Increased adiponectin receptor-1 expression in adipose tissue of impaired glucose-tolerant obese subjects during weight loss. Eur J Endocrinol 2006;155:161–165. 41 Huypens P: Adipokines regulate systemic insulin sensitivity in accordance to existing energy reserves. Med Hypoth 2007;69:161–165.
42 Bastard JP: Leptin over expressed in adipocytes increases TNF alpha production and down regulates production of adiponectin. Eur Cytokine Netw 2006;17:4–12. 43 Dagogo-Jack S, Tykodi G, Umamaheswaran I: Inhibition of cortisol biosynthesis decreases circulating leptin levels in obese humans. J Clin Endocrinol Metab 2005;90:5333–5335. 44 Hersong LG, Linneberg A: The link between the epidemic of obesity and allergic diseases: does obesity induce decreased immune tolerance? Allergy 2007;62:1205–1213. 45 Ladwig KH, Marten-Mittag B, Loewel H, et al: C-reactive protein, depressed mood, and the prediction of coronary heart disease in initially healthy men: results from the MONICA-KORA Augsburg Cohort Study. Eur Heart J 2005;26:2537–2542. 46 Osei K, Gaillard T, Schuster D: Plasma adiponectin levels in high-risk African-Americans with normal glucose tolerance, impaired glucose tolerance and type 2 diabetes. Obes Res 2005;13:179–185.
Brian E. Leonard, PhD, DSc Department of Pharmacology National University of Ireland Galway (Ireland) Tel. + 353 91 555 292, E-Mail
[email protected] Metabolic Syndrome and Schizophrenia
11
Thakore J, Leonard BE (eds): Metabolic Effects of Psychotropic Drugs. Mod Trends Pharmacopsychiatry. Basel, Karger, 2009, vol 26, pp 12–24
Insulin Resistance in Bipolar Women: Effects of Mood-Stabilizing Drugs Mytilee Vemuri ⭈ Pascale Stemmle ⭈ Bowen Jiang ⭈ Anna Morenkova ⭈ Natalie Rasgon Department of Psychiatry, Stanford University, Stanford, Calif., USA
Abstract Women with bipolar disorder (BD) may have unique risk factors for insulin resistance (IR). Specific periods in a woman’s reproductive timeline, specifically pregnancy and after the menopause, may represent times of increased IR. Moreover, women with BD demonstrate higher rates of obesity compared to men with BD, suggesting a sex-specific vulnerability to metabolic sequelae in BD. Additional contributors to metabolic sequelae, such as psychotropic medication, dysregulation of the hypothalamic-pituitary-adrenal axis, and genetic influences common in BD, may also manifest differently between the sexes. Several studies have suggested that women with BD may have more menstrual cycle irregularities than women in the general population. It has been hypothesized that such irregularities may be due to endocrinological disorders, such as polycystic ovarian syndrome or to hypothalamic-pituitary-adrenal axis dysfunction, both of which are also be associated with IR. Women treated with valproate may be particularly vulnerable to such irregularities. Lithium has not been observed to be associated with IR; however, it could theoretically influence IR through weight gain or effects of hypothyroidism. Treatment with atypical antipsychotics is associated with a greater risk of metabolic abnormalities, but sex-specific effects have not been observed. Definitive associations with IR have not thus far been demonstrated with either carbamazepine or lamotrigine. Copyright © 2009 S. Karger AG, Basel
High rates of metabolic disorders such as obesity, diabetes, lipid abnormalities and/ or metabolic syndrome (MetS) have been observed in patients with bipolar disorder (BD). Insulin resistance (IR) often underlies metabolic disorders and has been hypothesized to be either (1) an underlying state predisposing both mood disorders and metabolic disorders; (2) a side effect of medications used to treat mood disorders, and/or (3) a consequence of mood disorders themselves (due to lifestyle changes or stress resulting from the disorder). Women with BD may represent a group needing specific treatment considerations. Sex differences in IR, unique metabolic sequelae of mood stabilizing medications in women, and reproductive events that may alter insulin-sensitivity are all factors that merit consideration in women with BD.
In 1988, Reaven [1] proposed that the cluster of IR (and by definition, hyperinsulinemia), impaired glucose tolerance, abnormalities of plasma lipids, and hypertension were part of a single syndrome, syndrome X. It has also been called the insulin resistance syndrome and, more commonly, the MetS. Insulin resistance is defined as a reduced responsiveness of a target cell or a whole organism to the insulin concentration to which it is exposed [2]. Thus, IR can lead to reduced cellular uptake of glucose, hyperglycemia, and resultant increased insulin production from the pancreas.
Sex Differences in Insulin Resistance
Insulin resistance can vary between sexes and across a women’s reproductive life cycle. For example, some studies have suggested that girls through adolescence are inherently more insulin resistant than boys, but this relationship is thought to reverse after puberty [3, 4]. In adults, fat distribution patterns favor the development of IR in men, as men are more likely to develop abdominal obesity. Abdominal fat tissue is a major source of free fatty acids and cytokines and is associated with IR [5]. Premenopausal adult women more frequently develop peripheral obesity with subcutaneous fat accumulation, whereas men and postmenopausal women are more prone to central or abdominal obesity. After the menopause, concentrations of lipoproteins as well as body fat distribution shift to a more male pattern [5]. Pregnancy is also a time of elevated peripheral IR for women. During pregnancy, there is an approximate 50% reduction in insulin sensitivity by the third trimester; such changes are thought to be adaptive mechanisms to shunt nutrients to the developing fetus [6]. Glucose abnormalities can present differently between men and women. In a study of elderly people, the diagnosis of diabetes solely based on post-challenge hyperglycemia was more frequent in women than in men [7].
Individuals with Bipolar Disorder at Risk for Metabolic Disorders
The estimated prevalence of MetS or insulin resistance syndrome (IRS) in the USA is 24% in adults [8]. Many studies have demonstrated similarly high or higher rates of MetS in patients with BD, ranging from 16.7% to 49% [9–14]. Similarly, several retrospective studies have demonstrated an increased risk of diabetes in patients with BD [15–17]. Even patients without known diabetes or lipid abnormalities demonstrated high rates of diabetes (6.7%) and prediabetic glucose abnormalities (23.3%), including impaired fasting glucose and impaired glucose tolerance [10]. The majority of these BD patients were of normal weight, suggesting that metabolic abnormalities cannot entirely be explained by weight gain. Atypical antipsychotics such as olanzapine, risperidone and quetiapine are associated with the development or exacerbation of diabetes mellitus in bipolar patients [18]. However, one study has also shown
Insulin Resistance in Bipolar Women
13
medicated patients with BD are vulnerable to hyperinsulinemia, particularly those who gain weight or use beta-blockers, regardless of the type of psychotropic used [18a]. We reported high rates of IR in all of our studies in women with BD, regardless of the type of psychotropic medication used [19–22]. Metabolic dysfunction may also be present prior to psychotropic treatment – recent data from our group [22a] found high rates of metabolic dysfunction and obesity in a small sample of untreated women with BD. Therefore, additional factors such as concurrent therapy with other drugs, genetic influences, and the pathophysiological etiology of the BD illness itself may contribute to IR. Intrinsic neuroendocrine features of BD may lead to somatic metabolic dysfunction. In both the depressive and manic phases of BD, there is a positive association with chronic stress and elevated cortisol [23]. These abnormalities, along with frequent hyperactivity of the hypothalamic-pituitary-adrenal (HPA) axis, could potentially contribute to the higher prevalence of MetS. Chronically elevated glucocorticoids from HPA hyperactivation impedes glucose uptake by insulin, which in turn promotes metabolic dysfunction and cardiovascular disease [24]. Dysregulation of the HPA axis is also associated with obesity and elevated levels of leptin, a hormone that mediates central regulation of body mass, suggesting that the obesity may be due to inefficient leptin signaling and decreased feelings of satiety [23]. Another consequence of elevated cortisol is increased activity of lipoprotein lipase, which increases the amount of adipose tissue [25]. Elevated cortisol secretion also causes IR in muscles and affects the function of glycogen synthase, which can then promote peripheral IR [26, 27].
Specific Metabolic Features of Women with Bipolar Disorder
Metabolic Syndrome Baseline data from the Clinical Antipsychotic Trial of Intervention Effectiveness (CATIE) demonstrated higher rates of MetS in women with schizophrenia (51.6%), when compared with women in the general population and with men with schizophrenia (36.0%) [18]. However, gender differences in MetS prevalence in BD patients have not emerged as clearly. The majority of studies of prevalence rates of MetS in BD have not noted significant gender differences, but have involved smaller numbers than those in the CATIE study [12, 13]. One study demonstrated a higher prevalence of MetS in bipolar women in comparison with bipolar men [14].
Obesity Individuals with BD demonstrate higher rates of obesity than the general population [28]. Women with BD appear to have higher rates of obesity than men with BD [29],
14
Vemuri · Stemmle · Jiang · Morenkova · Rasgon
and a propensity towards higher waist circumferences [10]. Obese patients with either BD or schizophrenia are more likely to be women [30], and weight gain has been shown to be associated with female sex [31]. An evaluation of data from the Systematic Treatment Enhancement Program for Bipolar Disorder (STEP-BD) demonstrated greater rates of obesity in bipolar women (31%), compared to bipolar men (21%); in contrast, there were greater rates of overweight in bipolar men (38%), compared to women (22%) [32], a finding consistent with previous studies [33]. Women with BD appear to have a higher likelihood of increased waist circumference than men with BD, suggesting a sex-specific vulnerability to IR in BD. Changes in weight and appetite are found more commonly in women than in men with BD [34, 35]. Higher rates of thyroid and eating disorders are seen in women with BD [36]. Factors that influence the onset and maintenance of obesity in BD include both gender and eating behavior [37]. Similarly, both sex and eating disorders significantly correlate with overweight and obesity in BD [33]. Sex differences in comorbid thyroid disease, bulimia, and appetite changes could theoretically lead to more weight gain in bipolar women.
Menstrual Irregularities and Bipolar Disorder
Several studies have suggested that women with BD may have more menstrual cycle irregularities than women in the general population. It has been hypothesized that such irregularities are due to endocrinological disorders, specifically polycystic ovarian syndrome (PCOS) or due to HPA dysfunction, both of which could be associated with IR and metabolic dysfunction. PCOS is one of the most common endocrine disorders occurring in reproductiveaged endocrine disorder with an estimated prevalence of between 4 and 6% [38]. It is characterized by chronic anovulation and hyperandrogenism [39]. Chronic anovulation can lead to menstrual abnormalities and infertility. Hyperandrogenism can manifest as hirsutism (excess hair growth on the face), acne, and male-pattern balding. Metabolic consequences of PCOS include obesity and IR, which may lead to type-2 diabetes mellitus and cardiovascular disease (CVD) [39–42], and underscores the relationship between reproductive abnormalities and IR in women. PCOS is associated with overweight or obesity in approximately 50% of women with the disorder [43]. Even if they are normal weight, individuals with PCOS tend to have a higher proportion of abdominal body fat [44, 45], which is a risk factor for both IR [46] and CVD [47]. Lipid abnormalities have also been found at high rates among women with PCOS, including low high-density lipoprotein cholesterol (HDL-C) levels and elevated triglyceride levels [48, 49]. Women with PCOS may also exhibit increased immunological responses, such as elevated high sensitivity C-reactive protein (hsCRP) and interleukin-6 (IL-6), which can lead to subclinical CVD and impairment of endothelial structure [50–53]. Thus, the development of PCOS may present significant medical complications for many women.
Insulin Resistance in Bipolar Women
15
Mood Stabilizers and Impact on Insulin Resistance in Women with Bipolar Disorder
Women with BD are often treated with psychotropic agents, including mood stabilizers, atypical antipsychotics, and antidepressants, whose effects on reproductive function and IR are not fully understood. Medications may impact weight and endocrine abnormalities in a variety of ways that contribute to IR. For example, it has been theorized that valproate can influence the development of menstrual abnormalities by decreasing estrogen levels, increasing luteinizing hormone and increasing testosterone [54]. The increase in testosterone can lead to an arrest in maturation of ovarian follicles, leading to the development of polycystic ovaries. Additionally, valproate may cause an increase in leptin resulting in increased body weight [54], or lead to glucosestimulated insulin secretion by pancreatic cells [55]. Weight gain itself, not specific to valproate use, may lead to menstrual abnormalities or IR. In our studies, we demonstrated a correlation between BMI and both testosterone and insulin levels in women with BD [54]. Weight gain on psychotropic medications also predicted new onset menstrual abnormalities [54]. Thus, weight gain (whether it is inherent to BD or a result of any weight-liable mood stabilizer) may contribute to IR, particularly in women. Here, we review the evidence for the association between certain medications implicated in IR.
Valproate Valproate has been reported to have associations with PCOS in women with epilepsy and BD. Concerns over an association between valproate and PCOS first arose in valproate-treated women with epilepsy who were found to have high rates of PCOS [56]. As valproate is commonly used to treat women with BD, questions about its possible impact on endocrine and reproductive function in psychiatric populations emerged. Several cross-sectional studies have found increased rates of PCOS, menstrual abnormalities and/or hyperandrogenism in bipolar patients using valproate [22, 57, 58]. A study of 38 bipolar women [58] receiving either valproate or lithium monotherapy for at least 2 years demonstrated higher rates of menstrual abnormalities in patients receiving valproate (50%) than in patients receiving lithium (15%). Menstrual abnormalities were more prevalent in overweight/obese patients than in lean patients. Free testosterone and androstenedione levels were significantly higher than the reference range in valproate-treated patients, and luteinizing hormone was elevated in both groups. A similar study demonstrated that women receiving valproate had a significantly greater rate of menstrual abnormalities (47%) than women receiving non-valproate therapy (13%) and control women (0%) [57]. Forty-one percent of women with BD taking valproate had PCOS. A larger study of 300 women in the STEP-BD study found 10.6% (n = 9) of 86 patients treated with valproate had new onset menstrual cycle irregularities and hyperandrogenism within the first year of valproate use,
16
Vemuri · Stemmle · Jiang · Morenkova · Rasgon
most of of whom were also obese and had IR [59]. A follow-up study demonstrated reversal of PCOS reproductive features in 3 of 4 women upon discontinuation of valproate, while symptoms continued in 3 women continuing valproate [60]. Our own findings on this topic have yielded mixed results about the association of valproate with PCOS features; however we have consistently shown high rates of menstrual abnormalities in women with BD, even preceding diagnosis and treatment for the disorder. A pilot study of 22 women with BD, receiving lithium monotherapy, valproate monotherapy, or lithium-valproate combination therapy did not reveal a significant association between PCOS and valproate or lithium therapy [19]. All patients on lithium monotherapy or combination therapy and 60% of patients on valproate monotherapy reported menstrual disturbances. There were no significant differences in BMI or hirsutism, and hormone levels were within normal limits for all 3 groups. None of the subjects met NIH-defined criteria for PCOS. In a follow-up crosssectional study [22], we examined 80 reproductive-aged women who were receiving valproate therapy or non-valproate therapy for BD. Fifty-two of the 80 women (65%) reported current menstrual abnormalities, 40 of which (50%) reported one or more menstrual abnormalities that preceded the diagnosis of BD. Fifteen women (38%) reported developing menstrual abnormalities since beginning treatment for BD, 14 of whom developed abnormalities since treatment with valproate. No significant differences were observed between valproate and non-valproate groups in mean levels of free or total serum testosterone. Three of the 50 women (6%) taking valproate met the criteria for PCOS, compared to 0% of the 22 taking other mood-stabilizer medications. We conducted the first longitudinal study of this relationship, evaluating reproductive endocrine and metabolic markers in 25 women treated for BD over a 2-year time period [21]. At baseline, 10 (40%) women were currently undergoing treatment with valproate. Consistent with other published reports described above, 41.7% of subjects reported current oligomenorrhea, while 40% reported oligomenorrhea before starting medication. Rates of oligomenorrhea and clinical hyperandrogenism did not differ by type of medication use. Eighty percent of all subjects had a high homeostatic model assessment of IR (HOMA-IR) at baseline. Valproate use was associated with an increase over time in total testosterone. In contrast to other studies, we did not find co-occurrence of new-onset menstrual abnormalities with hyperandrogenism. We concluded that oligomenorrhea, hyperandrogenism, and IR are common in women with BD, and increases in androgen hormones may be related to valproate use [21].
Lithium Lithium could theoretically influence IR in women through weight gain or through effects of hypothyroidism. Lithium exposure increases the incidence of
Insulin Resistance in Bipolar Women
17
hypothyroidism. Clinical and subclinical hypothyroidism is more commonly seen in women than men with BD and in the general population, and the risk increases with age [61–63]. Hypothyroidism can cause alterations in lipid metabolism, weight gain, and menstrual cycle abnormalities, which in turn could impact IR. Lithium treatment in patients with BD has been associated with weight gain in numerous studies [64–68] and has been found to range from 5 kg within 1–2 years to 4.5–15.6 kg over 2 years [69, 70]. A recent animal study found that lithium increased gastrointestinal weight of male and female rats but only increased total body weight in females [71]. The mechanisms for lithium-induced weight gain remain unclear despite several conjectures. Lithium appears to exert insulin-like activity on carbohydrate metabolism, leading to increased glucose absorption in adipose tissue [72–75]. Lithium may also have direct effects on the hypothalamus to stimulate appetite and/or thirst and can also result in increased fluid retention [76]. Although lithium treatment can result in significant weight gain and/or hypothyroidism, long-term lithium treatment does not appear to be associated with IR or increased risk of developing diabetes mellitus [66]. In fact, one study suggested that lithium exerts anti-diabetic effects by lowering blood glucose levels in a manic-depressive female with adult onset diabetes mellitus [77].
Atypical Antipsychotics Atypical antipsychotic (AAP) treatment is also associated with a greater risk of metabolic abnormalities, such as dyslipidemia, weight gain, and new-onset type 2 diabetes mellitus [78]. Sex-specific impacts of atypical antipsychotics on IR have not been elucidated. Recent data suggest that AAPs may be more weight-liable medications than valproate [79]. In general, clozapine and olanzapine treatment are associated with the greatest risk of weight gain, while risperidone, quetiapine, ziprasidone, aripiprazole, amisulpride and zotepine have relatively lower levels of risk [80, 81]. In several studies examining IR via glucose tolerance testing, patients who received clozapine or olanzapine demonstrated higher degrees of resistance compared with those treated with risperidone [82–84]. Furthermore, in a recent study of 242 patients, patients treated with olanzapine or clozapine had significantly higher prevalence of dyslipidemia compared to unmedicated patients, independent of BMI [85]. A large matched case-control study of patients treated with AAPs conducted by Olfson et al. [86] found that treatment with clozapine, risperidone, quetiapine, olanzapine, ziprasidone, but not aripiprazole, was associated with a significant risk for hyperlipidemia, as compared to no AAP medication. There is clear correlation between obesity and IR, as adipocytes secrete several hormones (leptin, adipsin, adiponectin, tumor necrosis factor-α) that impair insulin secretion and/or insulin effects [87]. With the exception of adiponectin, all the hormones secreted by the endocrine adipose tissue exert antagonistic effects on insulin secretion and action [88]. As mentioned above, the use of AAPs is strongly associated with clinical weight gain and obesity; therefore, IR may be the result of increased
18
Vemuri · Stemmle · Jiang · Morenkova · Rasgon
adipose tissue and their hormone secretions. AAPs can also produce hyperinsulinemia through their antagonistic effects on dopamine (D2) receptors. D2 receptors are expressed in the β-cells and mediate glucose-stimulated secretion of insulin. Impaired dopamine receptor activation by via the effects of AAP may result in a compensatory increase in insulin secretion, which over time may develop into insulin insensitivity and resistance [89, 90]. Hormones important to appetite regulation and maintenance of adipose homeostasis such as leptin (anorectic) and ghrelin (orexigenic) may be disrupted by AAP treatment. It is hypothesized that AAP treatment disrupts appetite regulation by increasing ghrelin and/or decreasing leptin, thereby increasing Agoutirelated peptide (AGRP), neuropeptide Y (NP-Y), and orexin, leading to increased food intake and obesity. Ghrelin levels in patients treated with AAP for 1 year were significantly higher than in placebo-treated controls, with no difference between the various atypical agents that were used (clozapine, olanzapine, risperidone, or quetiapine) [91]. However, several other reports have indicated no difference in ghrelin levels or a reduction in ghrelin levels [92–94].
Carbamazepine, Lamotrigine Carbamazepine (CBZ) may be associated with weight gain and metabolic abnormalities, but to a lesser extent than valproate or lithium [95]. In a study of 105 epileptic women treated with valproate and CBZ monotherapy, rates of PCOs were similar (in both groups) to rates in the general population. However, BMI, triglycerides, and postprandial insulin were higher in valproate-treated patients than CBZ-treated patients. Of note, the enzyme-inducing properties of CBZ may reduce levels of free and total thyroxin in patients with thyroid dysfunction [96]. To date, the literature has not supported an association between lamotrigine (LTG) and IR. In a study of 54 women with epilepsy, MetS was more frequently associated with VPA-treated patients (41.7%) than CBZ (5.3%), LTG (0%), or topiramate (TPM) groups (0%) [97]. A post-hoc analysis was performed from a 12-week prospective open-label study of 1,175 patients with BD initiated on lamotrigine either as monotherapy or adjuvant treatment to valproate, lithium, antipsychotics, or antidepressants. No weight or BMI changes were noted after lamotrigine monotherapy or adjuvant therapy [98]. These findings are consistent with studies of lamotrigine in epilepsy populations [99].
Topiramate and Zonisamide While not approved for use as mood stabilizers, topiramate and zonisamide have potential roles in treating patients with BD. Both medications are considered weight neutral, and may possibly even contribute to weight loss. In patients with epilepsy and
Insulin Resistance in Bipolar Women
19
type 2 diabetes, patients showed better glycemic control when treated with topiramate [100]. Evidence has supported the utility of topiramate in treating binge-eating episodes in patients with obesity [101]. Both topiramate and zonisamide may have a role in promoting weight loss in patients with BD [102].
Conclusions
A growing literature supports an association between insulin resistance and bipolar disorder in women. Obesity in itself is a major contributor to IR and is a is highly prevalent in BD, more so in women with BD. Women with BD may be at risk for insulin resistance even prior to mood stabilizer initiation, and may also be susceptible to additional risks with certain mood stabilizers as weight gain associated with intake of these agents may further increase risk of insulin resistance. Mood stabilizing drugs confer variable metabolic risks. Valproate in particular has been implicated in development of IR as a component of PCOS. Valproate, lithium, and CBZ may cause weight gain, which can cause or worsen insulin resistance. Hypothyroidism, a side effect of lithium, may also be associated with weight gain and again is more prominent in women with BD than men. Many of the second-generation antipsychotics, including olanzapine, risperidone, and quetiapine, have been implicated in weight gain, and glucose and lipid abnormalities independent of weight gain. Mood stabilizers likely play only a partial role in the development of insulin resistance and metabolic disorders in women with BD, and likely interact with a number of other features including patient genetics, baseline weight, and psychiatric illness. Therefore, inquiry into an individual patient’s medical and family history, particularly diabetes, hyperlipidemia, hypertension, obesity, and metabolic side effects of previous medication, is a critical part of medication management. Longitudinal, prospective studies needed to further describe the interaction between mood disorders, insulin resistance and mood stabilizing drugs used in its treatment. Further research needs to delineate gender differences in the effects of medications used to treat psychiatric disorders, and ultimately to develop specialized treatment strategies for women with bipolar disorder. In the meantime, clinicians are charged with the task of reducing the medical burden of insulin resistance and metabolic disorders in patients with major mental illnesses.
References 1 Reaven GM: Banting lecture 1988: role of insulin resistance in human disease. Diabetes 1988;37:1595– 1607. 2 Shanik MH, Xu Y, Skrha J, Dankner R, Zick Y, Roth J: Insulin resistance and hyperinsulinemia: is hyperinsulinemia the cart or the horse? Diabetes Care 2008;31(suppl 2):S262–S268.
20
3 Moran A, Jacobs DRJ, Steinberger J, et al: Changes in insulin resistance and cardiovascular risk during adolescence: establishment of differential risk in males and females. Circulation 2008;117:2361– 2368.
Vemuri · Stemmle · Jiang · Morenkova · Rasgon
4 Travers SH, Jeffers BW, Bloch CA, Hill JO, Eckel RH: Gender and Tanner stage differences in body composition and insulin sensitivity in early pubertal children. J Clin Endocrinol Metab 1995;80:172– 178. 5 Regitz-Zagrosek V, Lehmkuhl E, Weickert MO: Gender differences in the metabolic syndrome and their role for cardiovascular disease. Clin Res Cardiol 2006;95:136–147. 6 Galerneau F, Inzucchi SE: Diabetes mellitus in pregnancy. Obstet Gynecol Clin North Am 2004;31:907– 933, xi-xii. 7 Barrett-Connor E, Ferrara A: Isolated postchallenge hyperglycemia and the risk of fatal cardiovascular disease in older women and men. The Rancho Bernardo Study. Diabetes Care 1998;21:1236–1239. 8 Ford ES, Giles WH, Dietz WH: Prevalence of the metabolic syndrome among US adults: findings from the third National Health and Nutrition Examination Survey. JAMA 2002;287:356–359. 9 Yumru M, Savas HA, Kurt E, et al: Atypical antipsychotics related metabolic syndrome in bipolar patients. J Affect Disord 2007;98:247–252. 10 Van Winkel RDHM, Hanssens L, Wampers M, Scheen A, Peuskens J: Prevalence of diabetes and the metabolic syndrome in a sample of patients with bipolar disorder. Bipolar Disorders 2008;10:342–348. 11 Fagiolini A, Frank E, Scott JA, Turkin S, Kupfer DJ: Metabolic syndrome in bipolar disorder: findings from the Bipolar Disorder Center for Pennsylvanians. Bipolar Disord 2005;7:424–430. 12 Cardenas J, Frye MA, Marusak SL, et al: Modal subcomponents of metabolic syndrome in patients with bipolar disorder. J Affect Disord 2008;106:91–97. 13 Garcia-Portilla MP, Saiz PA, Benabarre A, et al: The prevalence of metabolic syndrome in patients with bipolar disorder. J Affect Disord 2008;106:197–201. 14 Teixeira PJ, Rocha FL: The prevalence of metabolic syndrome among psychiatric inpatients in Brazil. Rev Bras Psiquiatr 2007;29:330–336. 15 Lilliker SL: Prevalence of diabetes in a manic-depressive population. Compr Psychiatry 1980;21: 270–275 16 Cassidy F, Ahearn E, Carroll BJ: Elevated frequency of diabetes mellitus in hospitalized manic-depressive patients. Am J Psychiatry 1999;156:1417–1420. 17 Regenold WT, Thapar RK, Marano C, Gavirneni S, Kondapavuluru PV: Increased prevalence of type 2 diabetes mellitus among psychiatric inpatients with bipolar I affective and schizoaffective disorders independent of psychotropic drug use. J Affect Disord 2002;70:19–26. 18 Guo JJ, Keck PE Jr, Corey-Lisle PK, et al: Risk of diabetes mellitus associated with atypical antipsychotic use among Medicaid patients with bipolar disorder: a nested case-control study. Pharmacotherapy 2007; 27:27–35.
Insulin Resistance in Bipolar Women
18a Tsai SY, Lee HC, Chen CC: Hyperinsulinaemia associated with beta-adrenoceptor antagonist in medicated bipolar patients during manic episode. Prog Neuropsychopharmacol Biol Psychiatry 2007; 31(5);1038–1043. 19 Rasgon NL, Altshuler LL, Gudeman D, et al: Medication status and polycystic ovary syndrome in women with bipolar disorder: a preliminary report. J Clin Psychiatry 2000;61:173–178. 20 Rasgon N, Bauer M, Glenn T, Elman S, Whybrow PC: Menstrual cycle related mood changes in women with bipolar disorder. Bipolar Disord 2003; 5:48–52. 21 Rasgon NL, Reynolds MF, Elman S, et al: Longitudinal evaluation of reproductive function in women treated for bipolar disorder. J Affect Disord 2005;89:217–225. 22 Rasgon NL, Altshuler LL, Fairbanks L, et al: Reproductive function and risk for PCOS in women treated for bipolar disorder. Bipolar Disord 2005;7:246–259. 22a Stemmle PG, Kenna HA, Wang PW, Hill SJ, Ketter TA, Rasgon NL: Insulin resistance and hyperlipidemia in women with bipolar disorder. J Psychiatr Res 2008, May 17. 23 Cassidy F, Ritchie J, Carroll B: Plasma dexamethasone concentration and cortisol response during manic episodes. Biol Psychiatry 1998;43:747–754. 24 Brindley D, Rolland Y: Possible connections between stress, diabetes, obesity, hypertension and altered lipoprotein metabolism that may result in atherosclerosis. Clin Sci (Lond) 1989;77:453–461. 25 Bjorntorp P: The regulation of adipose tissue distribution in humans. Int J Obes Relat Metab Disord 1996;20:291–302. 26 DeFronzo R, Ferrannini E: Insulin resistance: a multifaceted syndrome responsible for NIDDM, obesity, hypertension, dyslipidemia, and atherosclerotic cardiovascular disease. Diabetes 1991;14:173–194. 27 Bowden CL, Calabrese JR, McElroy SL, et al: A randomized, placebo-controlled 12-month trial of divalproex and lithium in treatment of outpatients with bipolar I disorder: Divalproex Maintenance Study Group. Arch Gen Psychiatry 2000;57:481– 489. 28 Elmslie JL, Silverstone JT, Mann JI, Williams SM, Romans SE: Prevalence of overweight and obesity in bipolar patients. J Clin Psychiatry 2000;61:179–184. 29 Arnold LM: Gender differences in bipolar disorder. Psychiatr Clin North Am 2003;26:595–620. 30 Kolotkin RL, Corey-Lisle PK, Crosby RD, et al: Impact of obesity on health-related quality of life in schizophrenia and bipolar disorder. Obesity (Silver Spring) 2008;16:749–754. 31 Taylor V, MacQueen G: Associations between bipolar disorder and metabolic syndrome: a review. J Clin Psychiatry 2006;67:1034–1041.
21
32 Wang PW, Sachs GS, Zarate CA, et al: Overweight and obesity in bipolar disorders. J Psychiatr Res 2006;40:762–764. 33 McElroy SL, Frye MA, Suppes T, et al: Correlates of overweight and obesity in 644 patients with bipolar disorder. J Clin Psychiatry 2002;63:207–213. 34 Kawa I, Carter JD, Joyce PR, et al: Gender differences in bipolar disorder: age of onset, course, comorbidity, and symptom presentation. Bipolar Disord 2005;7:119–125. 35 Benazzi F: Gender differences in bipolar II and unipolar depressed outpatients: a 557-case study. Ann Clin Psychiatry 1999;11:55–59. 36 Baldassano CF, Marangell LB, Gyulai L, et al: Gender differences in bipolar disorder: retrospective data from the first 500 STEP-BD participants. Bipolar Disord 2005;7:465–470. 37 Wildes JE, Marcus MD, Fagiolini A: Obesity in patients with bipolar disorder: a biopsychosocialbehavioral model. J Clin Psychiatry 2006;67:904–915. 38 Franks S: Polycystic ovary syndrome. N Engl J Med 1995;333:853–861. 39 Dunaif A, Thomas A: Current concepts in the polycystic ovary syndrome. Annu Rev Med 2001;52:401– 419. 40 Dockerty MB, Jackson RL: The Stein-Leventhal syndrome: analysis of 43 cases with special reference to association with endometrial carcinoma. Am J Obstet Gynecol 1957;73:161–173. 41 Carmina E, Lobo RA: Polycystic ovary syndrome (PCOS): arguably the most common endocrinopathy is associated with significant morbidity in women. J Clin Endocrinol Metab 1999;84:1897– 1899. 42 Hopkinson ZE, Sattar N, Fleming R, Greer IA: Polycystic ovarian syndrome: the metabolic syndrome comes to gynaecology. BMJ 1998;317:329– 332. 43 Pasquali R, Casimirri F: The impact of obesity on hyperandrogenism and polycystic ovary syndrome in premenopausal women. Clin Endocrinol (Oxf) 1993;39:1–16. 44 Kirchengast S, Huber J: Body composition characteristics and body fat distribution in lean women with polycystic ovary syndrome. Hum Reprod 2001;16:1255–1260. 45 Carmina E, Bucchieri S, Esposito A, et al: Abdominal fat quantity and distribution in women with polycystic ovary syndrome and extent of its relation to insulin resistance. J Clin Endocrinol Metab 2007;92:2500–2505. 46 Third Report of the National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III) final report. Circulation 2002;106:3143–3421.
22
47 Larsson B, Bengtsson C, Bjorntorp P, et al: Is abdominal body fat distribution a major explanation for the sex difference in the incidence of myocardial infarction? The study of men born in 1913 and the study of women, Goteborg, Sweden. Am J Epidemiol 1992;135:266–273. 48 Wild RA, Alaupovic P, Parker IJ: Lipid and apolipoprotein abnormalities in hirsute women. I. The association with insulin resistance. Am J Obstet Gynecol 1992;166:1191–1196; discussion 1196–1197. 49 Legro RS, Blanche P, Krauss RM, Lobo RA: Alterations in low-density lipoprotein and highdensity lipoprotein subclasses among Hispanic women with polycystic ovary syndrome: influence of insulin and genetic factors. Fertil Steril 1999;72:990–995. 50 Tiras MB, Yalcin R, Noyan V, et al: Alterations in cardiac flow parameters in patients with polycystic ovarian syndrome. Hum Reprod 1999;14:1949– 1952. 51 Paradisi G, Steinberg HO, Hempfling A, et al: Polycystic ovary syndrome is associated with endothelial dysfunction. Circulation 2001;103:1410– 1415. 52 Orio F Jr, Palomba S, Spinelli L, et al: The cardiovascular risk of young women with polycystic ovary syndrome: an observational, analytical, prospective case-control study. J Clin Endocrinol Metab 2004;89:3696–3701. 53 Vural B, Caliskan E, Turkoz E, Kilic T, Demirci A: Evaluation of metabolic syndrome frequency and premature carotid atherosclerosis in young women with polycystic ovary syndrome. Hum Reprod 2005;20:2409–2413. 54 Diamanti-Kandarakis E, Piperi C, Spina J, et al: Polycystic ovary syndrome: the influence of environmental and genetic factors. Hormones (Athens) 2006;5:17–34. 55 Luef G, Abraham I, Haslinger M, et al: Polycystic ovaries, obesity and insulin resistance in women with epilepsy: a comparative study of carbamazepine and valproic acid in 105 women. J Neurol 2002;249:835–841. 56 Isojarvi JI, Laatikainen TJ, Pakarinen AJ, Juntunen KT, Myllyla VV: Polycystic ovaries and hyperandrogenism in women taking valproate for epilepsy. N Engl J Med 1993;329:1383–1388. 57 O’Donovan C, Kusumakar V, Graves GR, Bird DC: Menstrual abnormalities and polycystic ovary syndrome in women taking valproate for bipolar mood disorder. J Clin Psychiatry 2002;63:322–330. 58 McIntyre RS, Mancini DA, McCann S, Srinivasan J, Kennedy SH: Valproate, bipolar disorder and polycystic ovarian syndrome. Bipolar Disord 2003;5:28– 35.
Vemuri · Stemmle · Jiang · Morenkova · Rasgon
59 Joffe H, Cohen LS, Suppes T, et al: Valproate is associated with new-onset oligoamenorrhea with hyperandrogenism in women with bipolar disorder. Biol Psychiatry 2006;59:1078–1086. 60 Joffe H, Cohen LS, Suppes T, et al: Longitudinal follow-up of reproductive and metabolic features of valproate-associated polycystic ovarian syndrome features: a preliminary report. Biol Psychiatry 2006; 60:1378–1381. 61 Sit D: Women and bipolar disorder across the life span. J Am Med Womens Assoc 2004;59:91–100. 62 Kupka RW, Nolen WA, Post RM, et al: High rate of autoimmune thyroiditis in bipolar disorder: lack of association with lithium exposure. Biol Psychiatry 2002;51:305–311. 63 Kusalic M, Engelsmann F: Effect of lithium maintenance therapy on thyroid and parathyroid function. J Psychiatry Neurosci 1999;24:227–233. 64 Peselow E, Dunner D, Fieve R: Lithium Carbonate and Weight Gain. J Affect Disorder 1980;2:303–310. 65 Bowden C, Calabrese J, McElroy S: A randomized, placebo-controlled 12-month trial of divalproex and lithium in treatment of outpatients with bipolar I disorder. Divalproex Maintenance Study Group. Arch Gen Psychiatry 2000;57:481–489. 66 Vestergaard P, Amdisen A, Schou M: Clinically significant side effects of lithium treatment. Acta Psychiatr Scand 1980;62:193–200. 67 Garland E, Remick R, Zis A: Weight gain with antidepressants and lithium. J Clin Psychopharmacology 1998;8:323–330. 68 Vendsborg P, Bach-Mortensen M, Rafaelson O: Fat cell number and weight gain in lithium treated patients. Acta Psychiatr Scand 1976a;53:355–359. 69 Aronne L, Segal K: Weight gain in the treatment of mood disorders. J Clin Psychiatry 2003;64(suppl 8):22–30. 70 Baptista T, Teneud L, Contreras Q: Lithium and body weight gain. Pharmacopsychiatry 1995;28:35–44. 71 Levine S, Saltzman A: Lithium increases gastrointestinal tract weight of male or female rats but it increases body weight only in females. Prog Neuropsychopharmacol Biol Psychiatry 2008;32:29– 33. 72 Vendsborg P, Rafaelson O: Lithium in man: effect on glucose intolerance and serum electrolytes. Acta Psychiatr Scand 1973;49:601–610. 73 Plenges P, Mellerup E, Rafaelson O: Lithium action on glycogen synthesis in rat brain, liver and diaphragm. J Psychiatr Res 1970;8:29–36. 74 Mellerup E, Thomson H, Bjorun N: Lithium, weight gain and serum insulin in manic-depressive patients. Acta Psychiatr Scand 1972;48:332–336. 75 van der Velde C, Gordon M: Manic-depressive illness, diabetes mellitus, and lithium carbonate. Arch Gen Psychiatry 1969;21:478–485.
Insulin Resistance in Bipolar Women
76 Vieweg W, Godleski L, Hundley P, Yank G: Lithium, polyuria and abnormal diurnal weight gain in psychosis. Acta Psychiatr Scand 1988;78:510–514. 77 Saran A: Antidiabetic Effects of Lithium. J Clin Psychiatry 1982;43:383–384. 78 Baranyi A, Yazdani R, Haas-Krammer A, Stepan A: Atypical antipsychotics and metabolic syndrome. Wien Med Wochenschr 2007;157:255–270. 79 Elmslie J, Mann J, Silverstone J: Determinants of overweight and obesity in patients with bipolar disorder. J Clin Psychiatry 2001;62:486–491. 80 Newcomer J: Second-generation (atypical) antipsychotics and metabolic effects. CNS Drugs 2005; 19(suppl 1):1–93. 81 Baptista T, De Mendoza S, Beaulieu S, Bermudez A, Martinez M: The metabolic syndrome during atypical antipsychotic drug treatment: mechanisms and management. Metab Syndr Relat Disord 2004;2:290– 307. 82 Newcomer J, Haupt D, Fucetola R, et al: Abnormalities in glucose regulation during antipsychotic treatment of schizophrenia. Arch Gen Psychiatry 2002;59:337–345. 83 Bergman R, Ader M: Atypical antipsychotics and glucose homeostasis. J Clin Psychiatry 2005;66:504– 514. 84 Albaugh VL, Henry CR, Bello NT, et al: Hormonal and metabolic effects of olanzapine and clozapine related to body weight in rodents. Obesity (Silver Spring) 2006;14:36–51. 85 Birkenaes A, Birkeland K, Engh J, Farden A: Dyslipidemia independent of body mass in antipsychotic-treated patients under real-life conditions. J Clin Psychopharmacology 2008;28:132–137. 86 Olfson M, Marcus S, Corey-Lisle P, Tuomari A, Hines P, L’Italien G: Hyperlipidemia following treatment with antipsychotic medications. Am J Psychiatry 2006;163:1821–1825. 87 Vestri H, Maianu L, Moellering D, Garvey W: Atypical antipsychotic drugs directly impair insulin action in adipocytes: effects on glucose transport, lipogenesis, and antilipolysis. Neuropsychopharmacology 2007;32:765–772. 88 Richards AA, Hickman IJ, Wang AY, et al: Olanzapine treatment is associated with reduced high molecular weight adiponectin in serum: a potential mechanism for olanzapine-induced insulin resistance in patients with schizophrenia. J Clin Psychopharmacol 2006;26:232–237. 89 Baptista T, Kin NM, Beaulieu S, de Baptista EA: Obesity and related metabolic abnormalities during antipsychotic drug administration: mechanisms, management and research perspectives. Pharmacopsychiatry 2002;35:205–219.
23
90 Rubi B, Ljubicić S, Pournourmohammadi S: Dopamine D2-like receptors are expressed in pancreatic beta cells and mediate inhibition of insulin secretion. J Biol Chem 2005;280:36824–36832. 91 Palik E, Birkas K, Faludi G: Correlation of serum ghrelin levels with body mass index and carbohydrate metabolism in patients treated with atypical antipsychotics. Diabetes Res Clin Pract 2005; 68(suppl 1):S60–S64. 92 Sporn A, Bobb A, Gogtay N: Hormonal correlates of clozapine-induced weight gain in psychotic children: an exploratory study. J Am Acad Child Adolesc Psychiatry 2005;44:925–933. 93 Togo T, Hasegawa K, Miura S: Serum ghrelin concentrations in patients receiving olanzapine or risperidone. Psychopharmacology 2004;172:230–232. 94 Theisen F, Gebhardt S, Bromel T: A prospective study of serum ghrelin levels in patients treated with clozapine. J Neural Transm 2005;112:1411–1416. 95 Wang PW, Ketter TA: Clinical use of carbamazepine for bipolar disorders. Expert Opin Pharmacother 2005;6:2887–2902. 96 Steinhoff BJ: Optimizing therapy of seizures in patients with endocrine disorders. Neurology 2006;67(suppl 4):S23–S27.
97 Kim JY, Lee HW: Metabolic and hormonal disturbances in women with epilepsy on antiepileptic drug monotherapy. Epilepsia 2007;48:1366–1370. 98 Zarzar MN, Graham J, Roberts J, Thompson T, Nanry K: Effectiveness and weight effects of openlabel lamotrigine with and without concomitant psychotropic medications in patients with bipolar I disorder. MedGenMed 2007;9:41. 99 Ben-Menachem E: Weight issues for people with epilepsy: a review. Epilepsia 2007;48(suppl 9):42– 45. 100 Khanna V, Arumugam S, Roy S, Mittra S, Bansal VS: Topiramate and type 2 diabetes: an old wine in a new bottle. Expert Opin Ther Targets 2008;12:81– 90. 101 Tata AL, Kockler DR: Topiramate for binge-eating disorder associated with obesity. Ann Pharmacother 2006;40:1993–1997. 102 Post RM, Altshuler LL, Frye MA, et al: New findings from the Bipolar Collaborative Network: clinical implications for therapeutics. Curr Psychiatry Rep 2006;8:489–497.
Mytilee P. Vemuri, MD, MBA Stanford University, Department of Psychiatry 401 Quarry Road Stanford, CA 94305 (USA) Tel. +1 650 725–1774, Fax +1 650 724–9900, E-mail
[email protected] 24
Vemuri · Stemmle · Jiang · Morenkova · Rasgon
Thakore J, Leonard BE (eds): Metabolic Effects of Psychotropic Drugs. Mod Trends Pharmacopsychiatry. Basel, Karger, 2009, vol 26, pp 25–46
Obesity and Mental Illness1 Leslie Citromea ⭈ Betty Vreelandb a
New York University School of Medicine, Department of Psychiatry, and the Nathan S. Kline Institute for Psychiatric Research, Orangeburg, N.Y.; bUniversity Behavioral HealthCare, the School of Nursing, and the Departments of Psychiatry and Family Medicine, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, Piscataway, N.J., USA
Abstract Obesity is one of the most common physical health care problems among persons with severe and persistent mental illness. Obesity is a complex multifactorial chronic disease that develops from the interaction between genotype and the environment. Among individuals with serious mental illnesses, an unhealthy lifestyle, as well as the effects of psychotropic medications, such as secondgeneration antipsychotics, can contribute to the development of this problem. Excess body weight increases the risk for many medical problems, including type 2 diabetes mellitus, coronary heart disease, osteoarthritis, hypertension, and gallbladder disease. Monitoring body weight is essential, and weight gain early in treatment can help predict those at high risk for substantial weight gain. Children, adolescents, and first-episode patients are at higher risk for weight gain. Lifestyle therapies and other non-pharmacological interventions have been shown to be effective in controlled clinical trials, but the evidence base for adjunctive medication strategies such as with orlistat, sibutramine, amantadine, nizatidine, metformin, topiramate, and others, is conflicting. Switching antipsychotic medication may or may not be clinically feasible, but can lead to a reduction in body weight. At the very least, a ‘small steps approach’ to managing weight should be offered to all patients who are Copyright © 2009 S. Karger AG, Basel started on second generation antipsychotics.
Obesity is a complex multifactorial chronic disease that develops from the interaction between genotype and the environment. Obesity in individuals with mental disorders such as schizophrenia has been attributed to various factors, including a sedentary lifestyle, poor nutritional choices or lack of access to healthy foods, the effects of both the mental disorder itself and the medications used to treat it, and lack of access to adequate preventative medical care. Excess body weight increases the risk for many medical problems, including type 2 diabetes mellitus, coronary heart disease, osteoarthritis, hypertension, and gallbladder disease [1]. Abdominal or visceral obesity is 1
Portions of this chapter have been adapted with kind permission from: Citrome L, Vreeland B: Schizophrenia, obesity, and antipsychotic medications: what can we do? Postgrad Med 2008;120(2):18–33.
particularly associated with increased risk for insulin resistance and/or the metabolic syndrome and for type 2 diabetes mellitus [2], and persons with schizophrenia have greater visceral adiposity than healthy individuals [3]. The clinical problem of obesity has become more apparent with the availability of second-generation, or ‘atypical’, antipsychotics. Their advantage over the older ‘neuroleptics’ have principally been in their lower propensity for extrapyramidal side effects, including tremor, rigidity, and akathisia [4]. However, one of the most troubling adverse effects of the second-generation antipsychotics is treatment-associated weight gain. The second-generation antipsychotics available today differ in their propensity for weight gain and the degree of weight change can also vary from patient to patient. Efficacy may also differ from drug to drug, and patient to patient, making medication selection and monitoring for weight gain a complex issue and can give rise to significant therapeutic dilemmas. Moreover, obesity can be an obstacle to adherence to medication, as evidenced by a mail survey of persons with schizophrenia where BMI status and subjective distress from weight gain were predictors of noncompliance [5]. This chapter addresses what the clinician can do to ameliorate the problem of overweight and obesity observed in patients with mental disorders, particularly schizophrenia. When using antipsychotics, especially those most associated with weight gain, ongoing monitoring is essential. Certain patients, such as children, adolescents, and those with their first episode of schizophrenia, are at higher risk for weight gain, even when using the second-generation antipsychotic medications that are ordinarily considered as ‘weight-neutral’. Early monitoring can identify early weight gainers; these patients are at significant risk for substantial weight gain. Lifestyle interventions are crucial when weight gain occurs and switching antipsychotic medication is not a viable option. Adjunctive medications for weight loss can be considered but randomized clinical trials for this intervention have generally not been encouraging.
Definitions
Body mass index (BMI), calculated as the quotient of body weight (kg) divided by the square of height (m), is commonly used to assess body weight. On-line calculators for BMI are readily available (for example, the website from the Centers for Disease Control and Prevention, http://www.cdc.gov/nccdphp/dnpa/bmi/calc-bmi). Overweight is defined as a BMI between 25 and 29.9 inclusive, and obesity is defined as having a BMI of 30 or greater. BMI-related health risks are well established, with health risk considered ‘high’ for BMI of 30–34.9, ‘very high’ for BMI 35–39.9, and ‘extremely high’ for BMI greater than 40 [6]. Alternate definitions have focused on waist-to-hip ratios or waist circumference used in the definitions for metabolic syndrome developed by the World Health Organization and the National Cholesterol Education Program Expert Panel on
26
Citrome · Vreeland
Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (NCEP), respectively [7]. Moreover, these definitions may not apply in all ethnic groups. For example, waist circumference for Asians is generally lower, requiring a lower cut-off of 90 cm in men (instead of 102 cm in the NCEP definition) or 80 cm in women (instead of 88 cm), when assessing the prevalence of central obesity [8]. In Japan, the definition of obesity is a BMI of at least 25, rather than the 30 used among Caucasians [9]. Quantitative measures of body fat have been developed, notably dual energy X-ray absorptiometry (DEXA) [10].
Epidemiology
Obesity has reached epidemic proportions in the general population. Current International Obesity Task Force estimates suggest that at least 1.1 billion adults are overweight including 312 million who are obese; this represents a doubling and trebling in less than two decades [11]. Among randomly selected middle-aged participants from 34 populations in 21 countries from the early 1980s to the mid-1990s, mean BMI as well as the prevalence of overweight increased in virtually all Western European countries, Australia, the USA, and China, and was related to increasing trends in total energy supply per capita [12]. Since 1980, the prevalence of obesity in Great Britain in adults has almost trebled with obesity prevalence in 2002 of 23% for men and 25% for women [13]. In the US, obesity prevalence among adults in 2003– 2004 was 32%, and the prevalence of extreme obesity (BMI of at least 40) was 2.8% in men and 6.9% in women [14]. Patients with schizophrenia have higher rates of obesity [15]. Among the patients participating in phase 1 of the randomized and double-blinded 18-month Clinical Antipsychotic Trials of Intervention Effectiveness schizophrenia study (CATIE), the mean BMI was 30 [16]. Moreover, waist circumference exceeded norms for 46% of all subjects (n = 1435), 37% of all men (n = 1059), 73% of all women (n = 376), 48% of all Caucasians (n = 858), 43% of all Blacks (n = 504), and 47% of all Hispanics (n = 167).
Contributory Factors
Overweight and obesity is related to a mismatch between energy intake, expenditure, and storage [17]. A sedentary lifestyle associated with increased intake of high caloric food will lead to increased weight. For example, about 7,800 kcal of chemical energy is contained in 1 kg of adipose tissue so that even a small increase in intake (such as 0.5 liters a day of a sugared carbonated beverage or 200 kcal) can lead to a significant amount of weight gain of 9 kg in one year if these additional calories are not expended but instead stored as fat. The availability of different foods and the amount consumed
Obesity and Mental Illness
27
Table 1. What the world eats (compiled from [18]) Country
Meat consumption (kg per person per year)
Obesity prevalence men, women , %
Life expectancy, men, women, years
Australia
107
21, 23
78, 83
Bhutan
3
5, 13
60, 62
Bosnia
21
14, 22
69, 76
Chad
14
0.3, 1
46, 49
China
52
1, 1.5
70, 73
Cuba
32
12, 21
75, 79
Ecuador
45
6, 15
68, 74
Egypt
22
22, 39
65, 69
France
101
Germany Great Britain
7, 6
76, 84
82 (30 in sausages)
20, 19
76, 82
80
19, 21
76, 81
Greenland
114
16, 22
64, 70
Guatemala
24
13, 25
63, 69
India
5
0.9, 1.1
60, 62
Italy
90
12, 12
77, 83
Japan
44
2, 2
78, 85
Kuwait
60
30, 49
76, 77
Mali
19
0.4, 3
44, 46
Mexico
59
20, 32
72, 77
109
5, 25
60, 66
31
1, 3
72, 65
Mongolia Philippines Poland
78
13, 18
71, 79
Turkey
19
11, 32
68, 72
125
32, 38
75, 80
USA
varies from country to country. Table 1 outlines meat consumption, prevalence of obesity, and life expectancy for 24 different countries [18]. Patients with mental disorders such as schizophrenia may make poor dietary choices [19]. In patients with an increased risk for central adiposity to begin with [3], this combination of poor diet and lack of physical activity is particularly problematic. Psychotropic medications such as antipsychotics [4], antidepressants [20], and anticonvulsants [21], have been associated with weight gain. It is not uncommon for these medications to be used in combination. Possible mechanisms for medication-associated weight gain include weight loss before drug treatment, food craving, alteration in resting metabolic rate, sedation/decreased physical activity, change in neurotransmitters (in general, alpha-adrenergic neurotransmission is thought to stimulate appetite, whereas beta-adrenergic, histaminergic, dopaminergic, and sero-
28
Citrome · Vreeland
Table 2. Weight change in phase 1 of the Clinical Antipsychotic Trials of Antipsychotic Effectiveness for Schizophrenia trial [26] Antipsychotic
Mean weight change kg/month
Median weight change kg/month
5th and 95th percentile weight change, kg/month
Olanzapine
+0.909
+0.363
–0.636, +4.32
Quetiapine
+0.227
+0.045
–2.00, +2.86
Risperidone
+0.182
Perphenazine
–0.091
–0.045
–2.23, +1.82
Ziprasidone
–0.136
–0.136
–2.41, +2.68
0
–2.09, +2.59
toninergic signal transduction confers satiety) and alteration of neuropeptides such as leptin and cytokines such as tumor necrosis factor [22].
Second-Generation Antipsychotics
Second-generation antipsychotics are a major source of concern regarding weight gain. Attempts have been made to identify specific receptor-binding profiles of antipsychotics to aid in the prediction of propensity for weight gain [23], however there is much individual variation, and most of the information available at present comes from clinical observations. In a comprehensive meta-analysis of weight change after 10 weeks of treatment at a standard dose of antipsychotics, mean increases in body weight were calculated for the different medications [24]. Clozapine and olanzapine had the largest weight gains with 4.45 and 4.15 kg, respectively. Risperidone was associated with a more modest 2.10 kg gain. Ziprasidone appeared essentially weight neutral with a mean gain of 0.04 kg. Insufficient data were available to evaluate quetiapine at 10 weeks, but subsequent studies have indicated that quetiapine also has a moderate to high propensity toward gain, particularly over the long term, as demonstrated in a prospective, naturalistic study [25]. Aripiprazole, the newest second-generation antipsychotic medication to become commercially available, is generally considered to be weight neutral. Examining mean weight change as observed in clinical trials is not always informative as to how often this actually will be encountered in clinical practice. There is large individual variability regarding weight gain, even with agents commonly thought of as being inexorably tied to obesity. In the CATIE study weight change per month of antipsychotic treatment was reported both in terms of mean change and in terms of median and range [26]. Table 2 demonstrates that although mean weight change followed the expected pattern, some patients randomized to olanzapine lost 0.64 kg per month, and some gained 4.3 kg per month. Patients randomized to weight neutral medications still show variability in weight change, with some patients randomized
Obesity and Mental Illness
29
to ziprasidone losing 2.4 kg per month and others gaining 2.7 lb per month. Weight gain greater than 7% from baseline to last observation occurred among 30% of the patients randomized to olanzapine, 16% for quetiapine, 14% for risperidone, 12% for perphenazine, and 7% for ziprasidone. Number needed to treat (NNT) to encounter one additional case of weight gain greater than 7% when using olanzapine versus perphenazine (a first-generation antipsychotic) was 6, versus quetiapine 8, versus risperidone 7, and versus ziprasidone 5 [27]. This means for every 5 patients randomized to olanzapine instead of ziprasidone, there was one additional case of a patient on olanzapine gaining greater than 7% of their initial body weight. Keeping in mind patients randomized to olanzapine had a longer mean time on medication than the other antipsychotics, and thus had a greater opportunity to accumulate weight, these single-digit NNTs nevertheless represent clinically significant differences [28]. Switching antipsychotics can lead to weight loss. Ultimately, there will need to be a favorable balance of benefit to risk [29]. Deciding whether to continue treatment with a particular antipsychotic or switch the patient to another can pose substantial dilemmas for the clinician. The choice to ‘switch or stay’ is a highly individualized decision. Evidence-based medicine philosophy states that relevant clinical trials can inform the clinician in making thoughtful individualized treatment decisions but there are no guarantees of weight gain or loss or drug efficacy. With these caveats, consideration should be given to switching to a more weight-neutral medication when encountering rapid weight gain with a particular antipsychotic. For example, in the CATIE study, of 61 patients who gained over 7% of their body weight in phase 1, 42% of ziprasidone-treated patients, 20% of risperidone-treated patients, 7% of quetiapine-treated patients, and 0% of olanzapine-treated patients lost over 7% of their body weight during phase 2 [30]. The NNT to encounter one additional case of weight loss greater than 7% when using ziprasidone versus olanzapine or quetiapine was 3, and versus risperidone 5 [29]. The findings from CATIE are consistent with earlier reports from open-label studies, such as a post-hoc analysis of a 20-week study where switching from olanzapine to risperidone resulted in a reduction of prevalence of metabolic syndrome in overweight or obese patients, including reductions in body weight and BMI [31]. In another report of 3 studies in which outpatients experiencing were switched to 6 weeks of open-label ziprasidone, patients switched from olanzapine experienced a mean weight loss of 1.76 kg, those switched from risperidone had a lesser reduction in weight (–0.86 kg), and those switched from first-generation antipsychotics had a non-significant increase (+0.27 kg) [32]. In an 8-week study where patients were switched from other antipsychotics to open-label aripiprazole (92% were receiving olanzapine or risperidone prior to the switch), the mean weight loss from baseline ranged from 1.3 kg to 1.7 kg, the incidence of weight loss of at least 7% of total body weight ranged from 7% to 15%, and the incidence of weight gain of at least 7% of total body weight ranged from 3% to 5%, depending on the switching technique [33]. Early weight gain may be predictive of future substantial weight gain. One can use the time course for weight gain with olanzapine for early identification of those at
30
Citrome · Vreeland
risk before substantial weight gain has occurred [34, 35]. Patients taking olanzapine who gain 5% or more of their body weight within the first 4–12 weeks of treatment and who are going to remain on olanzapine will require more active behavioral and/ or pharmacologic interventions than patients who gain weight more slowly or not at all. Although weight gain may plateau after 40 weeks or so, by that time one-half of patients receiving olanzapine will have gained up to 10 kg, and some substantially more than that [36]. The pattern of weight gain with quetiapine is also consistent with the idea that it can occur early (within 12 weeks) [37]. Within the approved range, antipsychotic dose does not appear to be predictive of weight gain for olanzapine [36] but there may be differences at higher-than-approved doses [38]. For risperidone, in a short (6-week) trial, higher doses were associated with more weight gain [39]. Data regarding a dose relationship with weight gain with quetiapine is conflicting [37, 40]. Low initial BMI (88 cm (women) or >102 cm (men) and ≥2 risk factors including cigarette smoking, hypertension, high low-density lipoprotein (LDL ≥160 mg/dl), low highdensity lipoprotein (HDL 27)
26
53
yes
–4.2 kg (vs. +1.0 kg)
Mauri [48]
psychoeducational program including diet and use of a pedometer
olanzapine (and an 24 increase in BMI of at least 7%)
33
yes
–4.5 kg (vs. no change without the intervention)
Alvarez-Jiménez [49]
early behavioral intervention including behavioral interventions, nutrition, and exercise
drug-naive first-episode psychosis patients started on olanzapine, risperidone, or haloperidol
12
61
yes
+4.1 kg (vs. +6.9 kg); number experiencing weight gain greater than 7% was 39.3% (vs. 78.8%)
Littrell [50]
intervention group with psycho-education classes focused on nutrition, exercise, and living a healthy lifestyle
switched to olanzapine at start of study
16 (26)
70
yes
–0.03 kg (vs. +4.3 kg)
Evans [51]
nutrition education sessions
olanzapine (started within 12 weeks)
26
51
yes
+2.0 kg (vs. +6.0 kg); number experiencing weight gain greater than 7% was 13% (vs. 64%)
Vreeland [52]a Menza [53]a
nutrition, exercise, and behavioral interventions
olanzapine, risperidone, clozapine, quetiapine
12 (52)
31 (51)
yes
–3.0 kg (vs +3.2 kg)
Ball [54]a
weight watchers and offered supervised exercise sessions
olanzapine (and who 10 had gained at least 7% of their pretreatment body weight)
11 (22)
maybe
–2.3 kg (vs. –0.2 kg), but this was only significant for men and not women
Brar [55]
behavioral treatment
risperidone (switched from olanzapine and with a BMI > 26)
14
72
maybe
–2.0 kg (vs. –1.1 kg) but not statistically significant
Kwon [56]
education, diet, and exercise management
olanzapine (and an increase in weight of at least 7%)
12
48
yes
–3.94 kg (vs. –1.48 kg)
Weber [57]
cognitive/behavioral group intervention, modified after the Diabetes Prevention Project program
olanzapine, risperidone, ziprasidone, quetiapine (and BMI of at least 25 and impaired glucose tolerance)
16
15
maybe
–2.4 kg (vs. –0.6 kg) but not statistically significant
Author
Active intervention
Antipsychotic received (and other qualifications)
Harmatz [46]
behavior modification in which money is lost for failure to lose weight or group therapy or diet only
Wu [47]
34
Study length weeks
Citrome · Vreeland
Table 3. Continued Author
Active intervention
Antipsychotic received (and other qualifications)
Khazaal [58]
cognitive and behavioural treatment
Brown [59]
Study length weeks
n
Helpful?
Weight change observed in intervention group (vs. control group)
olanzapine, risperidone, 24 clozapine, quetiapine, amisulpride, firstgeneration antipsychotic (and weight gain af at least 2 kg)
61
yes
–3.5 kg (vs. +1.7 kg)
six weekly health promotion sessions
not reported
6
28
maybe
–0.40 kg (vs. +1.11 kg)
McKibbin [60]
diabetes awareness and rehabilitation training
any (and age over 40 years and with diabetes mellitus)
24
64
yes
–2.3 kg (vs. +2.7 kg)
Rotatori [61]
behavioral self-control program
not reported
14
14
yes
–3.3 kg (vs. +2.5 kg)
a
These studies had a comparison group composed of matched controls, rather than a randomized design. The numbers in parentheses represent the total number of subjects reported for both the intervention and comparison groups.
(sponsored by Eli Lilly and Company) and available in two formats: the ‘personalized’ program and the ‘manualized’ program. The Solutions for Wellness personalized program was evaluated in a study of 7,188 patients with mental illness, 83% of who were obese or overweight [65]. The 6-month intervention was individualized and used diet, exercise and behavioral strategies. Patients were not required to meet any enrollment criteria, including diagnosis, treatment, weight, or risk for weight gain. The mean change in weight was –2.77 kg and the mean change in BMI was –0.93. In addition, there is also a Solutions for Wellness manualized program [66], which is a free, manualized, psychoeducational wellness program designed to inspire and assist persons with mental illness to choose healthier eating and physical activity patterns. In contrast to the personalized program, which is designed for individuals who are more independent and can benefit from a self-study program, the manualized program was designed to be implemented with the support of health professionals with both groups and individuals. In a quasi-experimental study with 70 patients with schizophrenia or schizoaffective disorder participating in a 4-month intervention which involved a weekly 1-hour psychoeducational intervention using Solutions for Wellness (ed. 2), there was a positive effect on preventing antipsychotic-inducted weight gain as compared to a group who received treatment as usual [50] (table 3). A ‘small steps approach’ to weight loss and maintenance may be particularly beneficial to people with serious mental illnesses. Unlike traditional obesity treatment, the
Obesity and Mental Illness
35
goal is prevention of weight gain and modest weight loss over a longer period of time. The approach encourages the use of small everyday changes in dietary and physical activity patterns that people can sustain over time. The small steps approach suggests that trying to achieve some combination of reduced energy intake and increased physical activity that equals approximately 100 kcal/day (such as eliminating one can of a sugared carbonated beverage a day or adding 2,000 steps or about a mile of walking) should prevent weight gain in many adults [67]. A small steps approach was utilized in the Healthy Living study and is incorporated into the Solutions for Wellness (ed. 3) manualized program. In summary, many of the nonpharmacologic interventions used in the general population may be applicable to adults with major mental disorders. The research on lifestyle and other nonpharmacologic interventions in adults with schizophrenia and schizoaffective disorders indicates that significant weight loss is possible. Programs such as ‘Healthy Living’ and ‘Solutions for Wellness’ show promise. Clinicians should provide nutrition and physical activity counseling when starting treatment with a second-generation antipsychotic. At a minimum a ‘small steps’ approach to counseling patients about lifestyle changes is recommended with referral to more intense programs such as Solutions for Wellness, or more specialized care, such as registered dieticians when ‘small steps’ fail. More randomized controlled clinical trials are needed in this area.
Pharmacological Interventions
When nonpharmacological interventions fail to control weight gain, and when switching to another antipsychotic has been unsuccessful or is not possible, adjunctive pharmacotherapy may be considered. Drugs that are currently approved by the FDA for the treatment of obesity include orlistat and sibutramine, both of which should be used cautiously with psychotropic medications. These medications should be used only in combination with appropriate diet, exercise, and behavioral programs. Orlistat is an enteric inhibitor of pancreatic lipase, thus lowering the absorption of dietary fat. Adverse effects include flatulence and steatorrhea if too much fat is consumed. Orlistat was tested for 16 weeks in a randomized, double-blind, placebocontrolled clinical trial in overweight or obese patients treated with clozapine or olanzapine (n = 63, diagnosis not reported) [68]. Adherence to a behavioral program or diet was not required to participate in the study. No statistically significant effect was observed in the whole population, but male patients experienced modest weight loss (–2.36 vs. +0.62 kg on placebo). Weight loss of at least 5% of baseline was observed in 16% of the patients receiving adjunctive orlistat versus 6% receiving placebo (NNT 11, not statistically significant). All 4 patients who discontinued because of diarrhea were receiving orlistat. There is a report that orlistat did not alter bioavailability of haloperidol, clozapine, clomipramine, desipramine, or carbamazepine in 8 patients,
36
Citrome · Vreeland
however the authors noted the possibility of decreased absorption of concomitantly administered drugs in some individuals, and recommended plasma level monitoring [69]. Sibutramine affects the reuptake of norepinephrine, serotonin and dopamine, and was originally thought to be a potential antidepressant compound, but was ultimately commercialized as a weight loss agent. Sibutramine was tested in a 12-week doubleblind, randomized, placebo-controlled study in 37 overweight and obese subjects taking olanzapine for schizophrenia or schizoaffective disorder [70]. For the first 8 weeks all subjects participated in weekly group sessions focused on nutrition and behavioral modification. Although the sibutramine group exhibited a mean increase in systolic blood pressure of 2.1 mm Hg, and presented with a higher rate of anticholinergic side effects and sleep disturbances, greater weight loss was observed at week 12 versus placebo (–3.8 vs. –0.8 kg). A similarly designed study with clozapine conducted among 21 patients by the same research group failed to demonstrate a therapeutic advantage for adjunctive sibutramine [71]. Because sibutramine affects serotonin and norepinephrine reuptake, these two studies [70, 71] excluded patients who received tricyclic, selective serotonin reuptake inhibitor, or monoamine oxidase inhibitor antidepressants in the prior month. When compared with adjunctive topiramate in a 24-week randomized open-label clinical trial, adjunctive sibutramine resulted in similar amounts of weight loss as adjunctive topiramate [72], but generalizability of that study to schizophrenia is limited as the subjects had bipolar disorder and only 35% were receiving antipsychotic medication. Rimonabant is a cannabinoid antagonist that acts to control appetite and can lead to weight reduction. The proposed indication was to be weight management in people with a BMI of 30, or with a BMI of 27 and at least one comorbid medical condition. However, the manufacturer withdrew its application to sell this agent in the USA amid concerns that it may increase suicidal thinking and depression [73]. There are no published reports of its use in patients with schizophrenia receiving antipsychotics. Several attempts have been made in managing weight gain with adjunctive therapy utilizing non-FDA approved uses of several classes of medications, including oral hypoglycemics (metformin), anticonvulsants (topiramate), histamine H-2 receptor antagonists (nizatidine, famotidine), antiparkinsonian and antiviral agents (amantadine), antidepressants (reboxetine, fluoxetine, fluvoxamine), and others [74, 75]. However, few randomized clinical trials exist that test these agents among overweight or obese patients with schizophrenia. Approaches include the prevention of excessive weight gain when starting an antipsychotic, or the loss of weight already gained after receiving an antipsychotic for some time. Most of the studies enrolled small numbers of subjects and have mainly focused on ameliorating the weight gain observed with olanzapine. The randomized double-blind studies of these adjunctive agents [76–101] are summarized in table 4. Results are generally inconsistent. Many interventions as tested do not show a clinically significant benefit. Moreover, all adjunctive
Obesity and Mental Illness
37
Table 4. Double-blind, randomized, clinical trials for non-FDA-approved adjunctive pharmacological interventions for weight gain among patients with schizophrenia Author
Study medication (dose or dose range, with reported frequency)
Antipsychotic received Study length (and other qualifications) weeks
n
Helpful? Weight change observed in intervention group (vs. control group)
Graham [77]
amantadine (up to 300 mg/day)
olanzapine (and weight gain of at least 5 lb)
12
21
yes
BMI –0.07 (vs. +1.24)
Deberdt [78]
amantadine (100-300 mg/day)
olanzapine (and weight gain of at least 5%)
16 (24)
125
maybe
–0.19 kg (vs. +1.28 kg) but results at 24 weeks were not statistically significantly different
Cavazzoni [79]
nizatidine (150 mg b.i.d. or 300 mg b.i.d.)
olanzapine (newly started)
16
175
no
difference was not statistically significant at 16 weeks
Atmaca [80]
nizatidine (150 mg b.i.d.)
olanzapine (and weight gain in the range of 2.6–10.8 kg)
8
35
yes
–4.5 kg (vs. +2.3 kg)
Atmaca [81]
nizatidine (150 mg b.i.d.)
quetiapine (and weight gain in the range of 2.3–7.2 kg)
8
28
no
difference was not statistically significant
Assunção [82]
nizatidine (300 mg BID)
olanzapine (and weight gain of at least 5% from baseline)
12
54
no
difference was not statistically significant
Poyurovsky [83]
famotidine (40 mg/day)
olanzapine (first-episode patients)
6
14
no
difference was not statistically significant
Baptista [76]
metformin (850 mg/day – 850 mg b.i.d.) with sibutramine (10 mg/ day – 10 mg b.i.d.)
olanzapine
12
28
no
difference was not statistically significant
Wu [88]
metformin (250 mg t.i.d.)
olanzapine (drug-naïve first-episode patients)
12
40
yes
+1.9 kg (vs. +6.87 kg); number experiencing weight gain greater than 7% was 16.7% (vs. 63.2%)
Wu [89]
metformin (250 mg t.i.d.) and/or lifestyle intervention
clozapine, olanzapine, risperidone, sulpiride (and weight gain of more than 10%)
12
128
yes
lifestyle-plusmetformin group BMI –1.8 (vs. metformin-alone group –1.2 vs. lifestyle-plusplacebo group –0.5 vs. placebo group +1.2)
38
Citrome · Vreeland
Table 4. Continued
Author
Study medication (dose or dose range, with reported frequency)
Antipsychotic received Study length (and other qualifications) weeks
n
Helpful? Weight change observed in intervention group (vs. control group)
Klein [90]
metformin (500 mg/day – 850 mg b.i.d.)
olanzapine, risperidone, or quetiapine (and weight gain of more than 10%; children ages 10-17 years)
16
39
yes
stable weight (vs. +0.31 kg/week)
Baptista [91]
metformin (850–1,700 mg/day)
olanzapine
14
40
no
difference was not statistically significant
Baptista [92]
metformin (850–2,550 mg/day)
olanzapine
12
80
no
difference was not statistically significant
Ko [84]
topiramate (50 mg b.i.d. or 100 mg b.i.d.)
risperidone, olanzapine, quetiapine, or clozapine (and BMI of at least 25)
12
66
yes
–5.35 kg for topiramate 200 mg/ day (vs. –1.68 kg for topiramate 100 mg/ day vs. –0.3 kg for placebo)
Kim [85]a
topiramate (50 mg b.i.d.)
olanzapine
12
60
yes
+2.66 kg (vs. +4.02 kg)
Nickel [86] Egger [87]
topiramate (250 mg/day)
olanzapine (only women who had gained at least 5 kg; psychosis or bipolar)
10
43
yes
–4.4 kg (vs. +1.2 kg); at the end of an open-label 18 month observation period, –9.4 kg (vs. +5.5 kg)
McElroy [72]a
topiramate (25–600 mg/day) or sibutramine (5–15 mg/day)
various (bipolar patients, of which only 16 were receiving antipsychotics, and BMI of at least 30, or at least 27 kg/m2 with concomitant obesityrelated risk factors or diseases)
24
46
maybe
patients randomized either to sibutramine or topiramate lost comparable amounts of weight (4.1 and 2.8 kg, respectively)
Poyurovsky [98]
reboxetine (2 mg b.i.d.)
olanzapine (first-episode patients)
6
60
yes
+3.31 kg (vs. +4.91 kg); number experiencing weight gain greater than 7% was 19.4% (vs. 46.4%)
Obesity and Mental Illness
39
Table 4. Continued
Author
Study medication (dose or dose range, with reported frequency)
Antipsychotic received Study length (and other qualifications) weeks
n
Helpful? Weight change observed in intervention group (vs. control group)
Poyurovsky [97]
reboxetine (2 mg b.i.d.)
olanzapine (first-episode patients)
6
26
yes
+2.5 kg (vs. +5.5 kg); number experiencing weight gain greater than 7% was 20% (vs. 70%)
Poyurovsky [94]
fluoxetine (20 mg/day)
olanzapine (first-episode patients)
8
30
no
difference was not statistically significant
Bustillo [95]
fluoxetine (20–60 mg/day)
olanzapine (and gained at least 3% of weight)
16
30
no
+3 kg (vs. +1.7 kg)
Lu [96]a
fluvoxamine (50 mg/day)
clozapine (250 mg/day or less or 600 mg/d or less)
12
68
maybe
+0.9 kg (vs. +3.2 kg)
Borovicka [93]
phenylpropanolamine (75 mg/day)
clozapine (and weight gain of more than 10%)
12
16
no
difference was not statistically significant
Goodall [99]
fenfluramine (15 mg b.i.d.)
depot fluphenazine, flupenthixol, or clopenthixol (and BMI of at least)
12
29
maybe
rate of weight loss was significantly greater in those taking D-fenfluramine among the completers
Modell [100]
dextroamphetamine (5 mg/day)
thioridazine, chlorpromazine
8
20
no
+0.11 kg/week (vs. +0.01 kg/week)
Ding [101]
ling gui zhu gan tang mixture
clozapine, chlorpromazine, 8 perphenazine, risperidone, sulpiride, trifluoperazine (and body weight exceeding 20% of standard)
100
yes
–4.5 kg (vs. +5.0 kg)
a
Although randomized, this study was not double-blind.
medications may pose a tolerability problem for some patients, for example topiramate may be associated with cognitive dulling which can complicate the successful treatment of a patient with schizophrenia who may already be experiencing cognitive dysfunction [102]. Adjunctive reboxetine may be useful in preventing weight gain in first-episode patients [97, 98], but is currently not available in the USA. There are some non-Western treatments that may be helpful [101]. The adjunctive agent that
40
Citrome · Vreeland
holds the most promise, at least for persons early on in their illness, is adjunctive metformin [88–92].
Bariatric Surgery
There are no published controlled trials for the use of surgical interventions for the treatment of obesity for patients with schizophrenia. However, there is a report of a case series of 5 patients with schizophrenia and morbid obesity and a comparison was made with 165 nonpsychotic patients who also underwent bariatric surgery during a 1-year period [103]. The median BMI was 54 and all had obesity-related comorbidities, and all patients had been previously treated unsuccessfully with conservative methods of weight reduction. Median percent excess weight loss at 6 months was comparable to that achieved in the control group: excess weight loss was 39.5% in the patients with schizophrenia versus 46.9% in the controls (difference not statistically significant). In severely obese individuals with schizophrenia who have failed other attempts to address their disease, a careful individualized risk-to-benefit assessment for bariatric surgery should be considered. While having a history of a mental illness should not prevent people from getting bariatric surgery, evaluation of an individuals’ preoperative psychiatric status may play an important role in maximizing successful postoperative outcomes [104].
Conclusions
Overweight and obese individuals with serious mental illnesses are at high risk for many medical problems, particularly cardiovascular morbidity and mortality. When added weight is concentrated viscerally, the expected result is insulin resistance and the eventual development of type 2 diabetes mellitus for those vulnerable individuals whose pancreas can no longer compensate by producing additional insulin. The accumulation of excess body weight among patients with schizophrenia is multi-factorial, including unhealthy dietary choices, sedentary lifestyles and lack of access to appropriate food and physical activity sources, and perhaps a genetic or an intrinsic disease-state vulnerability for abdominal adiposity. Psychotropic medication, in particular the second-generation antipsychotics, have been associated with weight gain. These agents differ in their propensity for weight gain, with clozapine and olanzapine having the greatest likelihood of being associated with weight gain, and ziprasidone and aripiprazole the least. Switching from one agent to another, when clinically feasible, may help ameliorate weight gain. However, children, adolescents, and patients in their first episode of psychosis are at high risk for weight gain even with agents that are considered ‘weight-neutral.’ Monitoring weight is essential in individuals with mental illness receiving medication treatment, and early intervention
Obesity and Mental Illness
41
to reduce weight or prevent further weight gain is crucial in order to avoid becoming overweight or obese. Prevention of weight gain is indicated in any patient with a BMI of ≥25, and weight loss for any patient who has a BMI of ≥30 or a BMI of 25–29.9 and concomitant risk factors (such as hypertension, dyslipdemia, cardiovascular disease, diabetes, or sleep apnea [105]). Lifestyle and other nonpharmacological approaches show greater promise than using adjunctive medication treatments for weight loss, and the use of the latter requires nonpharmacological approaches to be in place in any event. The ‘small steps approach’ of setting modest nutritional and physical activity goals using accessible and realistic methods, is achievable during the routine care of individuals with serious mental illnesses.
References 1
2
3
4
5
6 7 8
9
42
Must A, Spadano J, Coakley EH, Field AE, Colditz G, Dietz WH: The disease burden associated with overweight and obesity. JAMA 1999;282:1523– 1529. Lois K, Young J, Kumar S: Obesity: eiphenomenon or cause of metabolic syndrome? Int J Clin Pract 2008;62:932–938. Thakore JH, Mann JN, Vlahos I, Martin A, Reznek R: Increased visceral fat distribution in drug-naive and drug-free patients with schizophrenia. Int J Obes Relat Metab Disord 2002;26:137–141. Citrome L, Volavka J: Atypical antipsychotics: revolutionary or incremental advance? Expert Rev Neurother 2002;2:69–88. Weiden PJ, Mackell JA, McDonnell DD: Obesity as a risk factor for antipsychotic noncompliance. Schizophr Res 2004;66:51–57. Bray GA: Evaluation of obesity. Postgrad Med 2003;114(6):19–38. Citrome L: Metabolic syndrome and cardiovascular disease. J Psychopharmacol 2005;19(suppl):84–93. Tan CE, Ma S, Wai D, Chew SK, Tai ES: Can we apply the National Cholesterol Education Program Adult Treatment Panel definition of the metabolic syndrome to Asians? Diabetes Care 2004;27:1182– 1186. Anuurad E, Shiwaku K, Nogi A, Kitajima K, Enkhmaa B, Shimono K, Yamane Y: The new BMI criteria for Asians by the regional office for the western pacific region of WHO are suitable for screening of overweight to prevent metabolic syndrome in elder Japanese workers. J Occup Health 2003;45:335–343.
10 Moyad MA: Fad diets and obesity. 1. Measuring weight in a clinical setting. Urol Nurs 2004;24:114– 119. 11 James PT, Rigby N, Leach R, International Obesity Task Forc: The obesity epidemic, metabolic syndrome and future prevention strategies. Eur J Cardiovasc Prev Rehabil. 2004;11:3–8. 12 Silventoinen K, Sans S, Tolonen H, Monterde D, Kuulasmaa K, Kesteloot H, Tuomilehto J: Trends in obesity and energy supply in the WHO MONICA Project. Int J Obes Relat Metab Disord 2004;28:710– 718. 13 Rennie KL, Jebb SA: Prevalence of obesity in Great Britain. Obes Rev 2005;6:11–12. 14 Ogden CL, Carroll MD, Curtin LR, McDowell MA, Tabak CJ, Flegal KM: Prevalence of overweight and obesity in the United States, 1999–2004. JAMA 2006; 295:1549–1555. 15 Allison DB, Fontaine KR, Heo M, Mentore JL, Cappelleri JC, Chandler LP, Weiden PJ, Cheskin LJ: The distribution of body mass index among individuals with and without schizophrenia. J Clin Psychiatry 1999;60:215–220. 16 McEvoy JP, Meyer JM, Goff DC, Nasrallah HA, Davis SM, Sullivan L, Meltzer HY, Hsiao J, Scott Stroup T, Lieberman JA: Prevalence of the metabolic syndrome in patients with schizophrenia: baseline results from the Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE) schizophrenia trial and comparison with national estimates from NHANES III. Schizophr Res 2005; 80:19–32. 17 Rosenbaum M, Leibel RL, Hirsch J: Obesity. N Engl J Med 1997;337:396–407. 18 Menzel P, D’Aluisio F: Hungry Planet. Napa, Material World Books, 2005.
Citrome · Vreeland
19 McCreadie RG, Kelly C, Connolly M, Williams S, Baxter G, Lean M, Paterson JR: Dietary improvement in people with schizophrenia: randomised controlled trial. Br J Psychiatry 2005;187:346–351. 20 Fava M, Judge R, Hoog SL, Nilsson ME, Koke SC: Fluoxetine versus sertraline and paroxetine in major depressive disorder: changes in weight with longterm treatment. J Clin Psychiatry 2000;61:863–867. 21 Vanina Y, Podolskaya A, Sedky K, Shahab H, Siddiqui A, Munshi F, Lippmann S: Body weight changes associated with psychopharmacology. Psychiatr Serv 2002;53:842–847. 22 Zimmermann U, Kraus T, Himmerich H, Schuld A, Pollmächer T: Epidemiology, implications and mechanisms underlying drug-induced weight gain in psychiatric patients. J Psychiatr Res 2003;37:193–220. 23 Matsui-Sakata A, Ohtani H, Sawada Y: Receptor occupancy-based analysis of the contributions of various receptors to antipsychotics-induced weight gain and diabetes mellitus. Drug Metab Pharmacokinet 2005;20:368–378. 24 Allison DB, Mentore JL, Heo M, Chandler LP, Cappelleri JC, Infante MC, Weiden PJ: Antipsychoticinduced weight gain: a comprehensive research synthesis. Am J Psychiatry 1999;156:1686–1696. 25 McIntyre RS, Trakas K, Lin D, Balshaw R, Hwang P, Robinson K, Eggleston A: Risk of weight gain associated with antipsychotic treatment: results from the Canadian National Outcomes Measurement Study in Schizophrenia. Can J Psychiatry 2003;48: 689–694. 26 Lieberman JA, Stroup TS, McEvoy JP, Swartz MS, Rosenheck RA, Perkins DO, Keefe RS, Davis SM, Davis CE, Lebowitz BD, Severe J, Hsiao JK, Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE) Investigators: Effectiveness of antipsychotic drugs in patients with chronic schizophrenia. N Engl J Med 2005;353:1209–1223. 27 Citrome L, Stroup TS: Schizophrenia, Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE) and number needed to treat: how can CATIE inform clinicians? Int J Clin Pract 2006;60: 933–940. 28 Citrome L: Compelling or irrelevant? Using number needed to treat can help decide. Acta Psychiatr Scand 2008;117:412–419. 29 Citrome L: The effectiveness criterion: balancing efficacy against the risk of weight gain. J Clin Psychiatry 2007;68(suppl 12):12–17. 30 Stroup TS, Lieberman JA, McEvoy JP, Swartz MS, Davis SM, Rosenheck RA, Perkins DO, Keefe RS, Davis CE, Severe J, Hsiao JK, CATIE Investigators: Effectiveness of olanzapine, quetiapine, risperidone, and ziprasidone in patients with chronic schizophrenia following discontinuation of a previous atypical antipsychotic. Am J Psychiatry 2006;163: 611–622.
Obesity and Mental Illness
31 Meyer JM, Pandina G, Bossie CA, Turkoz I, Greenspan A: Effects of switching from olanzapine to risperidone on the prevalence of the metabolic syndrome in overweight or obese patients with schizophrenia or schizoaffective disorder: analysis of a multicenter, rater-blinded, open-label study. Clin Ther 2005;27:1930–1941. 32 Weiden PJ, Daniel DG, Simpson G, Romano SJ: Improvement in indices of health status in outpatients with schizophrenia switched to ziprasidone. J Clin Psychopharmacol 2003;23:595–600. 33 Casey DE, Carson WH, Saha AR, Liebeskind A, Ali MW, Jody D, Ingenito GG, Aripiprazole Study Group: Switching patients to aripiprazole from other antipsychotic agents: a multicenter randomized study. Psychopharmacology (Berl) 2003;166: 391–399. 34 Kinon BJ, Kaiser CJ, Ahmed S, Rotelli MD, KollackWalker S: Association between early and rapid weight gain and change in weight over one year of olanzapine therapy in patients with schizophrenia and related disorders. J Clin Psychopharmacol 2005; 25:255–258. 35 Lipkovich I, Citrome L, Perlis R, Deberdt W, Houston JP, Ahl J, Hardy T: Early predictors of substantial weight gain in bipolar patients treated with olanzapine. J Clin Psychopharmacol 2006;26:316– 320. 36 Kinon BJ, Basson BR, Gilmore JA, Tollefson GD: Long-term olanzapine treatment: weight change and weight-related health factors in schizophrenia. J Clin Psychiatry 2001;62:92–100. 37 Brecher M, Leong RW, Stening G, OsterlingKoskinen L, Jones AM: Quetiapine and long-term weight change: a comprehensive data review of patients with schizophrenia. J Clin Psychiatry 2007; 68:597–603. 38 Kinon BJ, Volavka J, Stauffer V, Edwards SE, LiuSeifert H, Chen L, Adams DH, Lindemayer JP, McEvoy JP, Buckley PF, Lieberman JA, Meltzer HY, Eilson DR, Citrome L: Standard and higher dose of olanzapine in patients with schizophrenia or schizoaffective disorder: a randomized, double-blind, fixed dose study. J Clin Psychopharmacol 2008;28: 392–400. 39 Lane HY, Chang YC, Cheng YC, Liu GC, Lin XR, Chang WH: Effects of patient demographics, risperidone dosage, and clinical outcome on body weight in acutely exacerbated schizophrenia. J Clin Psychiatry 2003;64:316–320. 40 Arvanitis LA, Miller BG: Multiple fixed doses of ‘Seroquel’ (quetiapine) in patients with acute exacerbation of schizophrenia: a comparison with haloperidol and placebo: the Seroquel Trial 13 Study Group. Biol Psychiatry 1997;42:233–246.
43
41 Correll CU: Weight gain and metabolic effects of mood stabilizers and antipsychotics in pediatric bipolar disorder: a systematic review and pooled analysis of short-term trials. J Am Acad Child Adolesc Psychiatry 2007;46:687–700. 42 Strassnig M, Miewald J, Keshavan M, Ganguli R: Weight gain in newly diagnosed first-episode psychosis patients and healthy comparisons: one-year analysis. Schizophr Res 2007;93:90–98. 43 McEvoy JP, Lieberman JA, Perkins DO, Hamer RM, Gu H, Lazarus A, Sweitzer D, Olexy C, Weiden P, Strakowski SD: Efficacy and tolerability of olanzapine, quetiapine, and risperidone in the treatment of early psychosis: a randomized, double-blind 52-week comparison. Am J Psychiatry 2007;164: 1050–1060. 44 Kahn RS, Fleischhacker WW, Boter H, Davidson M, Vergouwe Y, Keet IP, Gheorghe MD, Rybakowski JK, Galderisi S, Libiger J, Hummer M, Dollfus S, López-Ibor JJ, Hranov LG, Gaebel W, Peuskens J, Lindefors N, Riecher-Rössler A, Grobbee DE, EUFEST Study Group: Effectiveness of antipsychotic drugs in first-episode schizophrenia and schizophreniform disorder: an open randomised clinical trial. Lancet 2008;371:1085–1097. 45 American Diabetes Association, American Psychiatric Association, American Association of Clinical Endocrinologists, North American Association for the Study of Obesity: Consensus Development Conference on Antipsychotic Drugs and Obesity and Diabetes. Diabetes Care 2004;27: 596–601. 46 Harmatz MG, Lapuc P: Behavior modification of overeating in a psychiatric population. J Consult Clin Psychol 1968;32:583–587. 47 Wu MK, Wang CK, Bai YM, Huang CY, Lee SD: Outcomes of obese, clozapine-treated inpatients with schizophrenia placed on a six-month diet and physical activity program. Psychiatr Serv 2007;58: 544–550. 48 Mauri M, Simoncini M, Castrogiovanni S, Iovieno N, Cecconi D, Dell’Agnello G, Quadrigli M, Rossi A, Donda P, Fagiolini A, Cassano GB: A psychoeducational program for weight loss in patients who have experienced weight gain during antipsychotic treatment with olanzapine. Pharmacopsychiatry 2008;41: 17–23. 49 Alvarez-Jiménez M, González-Blanch C, VázquezBarquero JL, Pérez-Iglesias R, Martínez-García O, Pérez-Pardal T, Ramírez-Bonilla ML, Crespo-Facorro B: Attenuation of antipsychotic-induced weight gain with early behavioral intervention in drug-naive firstepisode psychosis patients: A randomized controlled trial. J Clin Psychiatry 2006;67: 1253–1260. 50 Littrell KH, Hilligoss NM, Kirshner CD, Petty RG, Johnson CG: The effects of an educational intervention on antipsychotic-induced weight gain. J Nurs Scholarsh 2003;35:237–241.
44
51 Evans S, Newton R, Higgins S: Nutritional intervention to prevent weight gain in patients commenced on olanzapine: a randomized controlled trial. Aust N Z J Psychiatry 2005;39:479–486. 52 Vreeland B, Minsky S, Menza M, Rigassio Radler D, Roemheld-Hamm B, Stern R: A program for managing weight gain associated with atypical antipsychotics. Psychiatr Serv 2003;54:1155–1157. 53 Menza M, Vreeland B, Minsky S, Gara M, Radler DR, Sakowitz M: Managing atypical antipsychoticassociated weight gain: 12-month data on a multimodal weight control program. J Clin Psychiatry 2004;65:471–477. 54 Ball MP, Coons VB, Buchanan RW: A program for treating olanzapine-related weight gain. Psychiatr Serv 2001;52:967–969. 55 Brar JS, Ganguli R, Pandina G, Turkoz I, Berry S, Mahmoud R: Effects of behavioral therapy on weight loss in overweight and obese patients with schizophrenia or schizoaffective disorder. J Clin Psychiatry 2005;66:205–212. 56 Kwon JS, Choi JS, Bahk WM, Yoon Kim C, Hyung Kim C, Chul Shin Y, Park BJ, Geun Oh C: Weight management program for treatment-emergent weight gain in olanzapine-treated patients with schizophrenia or schizoaffective disorder: a 12-week randomized controlled clinical trial. J Clin Psychiatry 2006;67:547–553. 57 Weber M, Wyne K: A cognitive/behavioral group intervention for weight loss in patients treated with atypical antipsychotics. Schizophr Res 2006;83:95–101. 58 Khazaal Y, Fresard E, Rabia S, Chatton A, Rothen S, Pomini V, Grasset F, Borgeat F, Zullino D: Cognitive behavioural therapy for weight gain associated with antipsychotic drugs. Schizophr Res 2007;91:169–177. 59 Brown S, Chan K: A randomized controlled trial of a brief health promotion intervention in a population with serious mental illness. J Ment Health 2006;15:543–549. 60 McKibbin CL, Patterson TL, Norman G, Patrick K, Jin H, Roesch S, Mudaliar S, Barrio C, O’Hanlon K, Griver K, Sirkin A, Jeste DV: A lifestyle intervention for older schizophrenia patients with diabetes mellitus: a randomized controlled trial. Schizophr Res 2006;86:36–44. 61 Rotatori AF, Fox R, Wicks A: Weight loss with psychiatric residents in a behavioral self control program. Psychol Rep 1980;46:483–486. 62 National Heart Lung and Blood Institute, North American Association for the Study of Obesity: The Practical Guide: Identification, Evaluation, and Treatment of Overweight and Obesity in Adults. Bethesda, National Institutes of Health, 2000. 63 Tsai AG, Wadden TA, Womble LG, Byrne KJ: Commercial and self-help programs for weight control. Psychiatr Clin North Am 2005;28:171–192.
Citrome · Vreeland
64 Dansinger ML, Gleason JA, Griffith JL, Selker HP, Schaefer EJ: Comparison of the Atkins, Ornish, Weight Watchers, and Zone diets for weight loss and heart disease risk reduction: a randomized trial. JAMA 2005;293:43–53. 65 Hoffmann VP, Ahl J, Meyers A, Schuh L, Shults KS, Collins DM, Jensen L: Wellness intervention for patients with serious and persistent mental illness. J Clin Psychiatry 2005;66:1576–1579. 66 Vreeland B, Toto AM, Sakowitz M: Solutions for Wellness, ed 3. Indianapolis, Eli Lilly, 2008. 67 Hill JO, Wyatt HR: Small changes: a big idea for addressing obesity. Obesity Mgmt 2006;2:227–231. 68 Joffe G, Takala P, Tchoukhine E, Hakko H, Raidma M, Putkonen H, Eronen M, Räsänen P: Orlistat in clozapine- or olanzapine-treated patients with overweight or obesity: A 16-week randomized, doubleblind, placebo-controlled trial. J Clin Psychiatry 2008;69:706–711. 69 Hilger E, Quiner S, Ginzel I, Walter H, Saria L, Barnas C: The effect of orlistat on plasma levels of psychotropic drugs in patients with long-term psychopharmacotherapy. J Clin Psychopharmacol 2002; 22:68–70. 70 Henderson DC, Copeland PM, Daley TB, Borba CP, Cather C, Nguyen DD, Louie PM, Evins AE, Freudenreich O, Hayden D, Goff DC: A doubleblind, placebo-controlled trial of sibutramine for olanzapine-associated weight gain. Am J Psychiatry 2005;162:954–962. 71 Henderson DC, Fan X, Copeland PM, Borba CP, Daley TB, Nguyen DD, Zhang H, Hayden D, Freudenreich O, Cather C, Evins AE, Goff DC: A double-blind, placebo-controlled trial of sibutramine for clozapine-associated weight gain. Acta Psychiatr Scand 2007;115:101–105. 72 McElroy SL, Frye MA, Altshuler LL, Suppes T, Hellemann G, Black D, Mintz J, Kupka R, Nolen W, Leverich GS, Denicoff KD, Post RM, Keck PE Jr: A 24-week, randomized, controlled trial of adjunctive sibutramine versus topiramate in the treatment of weight gain in overweight or obese patients with bipolar disorders. Bipolar Disord 2007;9:426–434. 73 Christensen R, Kristensen PK, Bartels EM, Bliddal H, Astrup A: Efficacy and safety of the weight-loss drug rimonabant: a meta-analysis of randomised trials. Lancet 2007;370:1706–1713. 74 Faulkner G, Cohn T, Remington G: Interventions to reduce weight gain in schizophrenia. Schizophr Bull 2007;33:654–656. 75 Faulkner G, Cohn T, Remington G: Interventions to reduce weight gain in schizophrenia. Cochrane Database Syst Rev 2007;1:CD005148.
Obesity and Mental Illness
76 Baptista T, Uzcátegui E, Rangel N, El Fakih Y, Galeazzi T, Beaulieu S, de Baptista EA: Metformin plus sibutramine for olanzapine-associated weight gain and metabolic dysfunction in schizophrenia: a 12-week double-blind, placebo-controlled pilot study. Psychiatry Res 2008;159:250–253. 77 Graham KA, Gu H, Lieberman JA, Harp JB, Perkins DO: Double-blind, placebo-controlled investigation of amantadine for weight loss in subjects who gained weight with olanzapine. Am J Psychiatry 2005;162:1744–1746. 78 Deberdt W, Winokur A, Cavazzoni PA, Trzaskoma QN, Carlson CD, Bymaster FP, Wiener K, Floris M, Breier A: Amantadine for weight gain associated with olanzapine treatment. Eur Neuropsychopharmacol 2005;15:13–21. 79 Cavazzoni P, Tanaka Y, Roychowdhury SM, Breier A, Allison DB: Nizatidine for prevention of weight gain with olanzapine: a double-blind placebo-controlled trial. Eur Neuropsychopharmacol 2003;13: 81–85. 80 Atmaca M, Kuloglu M, Tezcan E, Ustundag B: Nizatidine treatment and its relationship with leptin levels in patients with olanzapine-induced weight gain. Hum Psychopharmacol 2003;18:457–461. 81 Atmaca M, Kuloglu M, Tezcan E, Ustundag B, Kilic N: Nizatidine for the treatment of patients with quetiapine-induced weight gain. Hum Psychopharmacol 2004;19:37–40. 82 Assunção SS, Ruschel SI, Rosa Lde C, Campos JA, Alves MJ, Bracco OL, de Lima MS: Weight gain management in patients with schizophrenia during treatment with olanzapine in association with nizatidine. Rev Bras Psiquiatr 2006;28:270–276. 83 Poyurovsky M, Tal V, Maayan R, Gil-Ad I, Fuchs C, Weizman A: The effect of famotidine addition on olanzapine-induced weight gain in first-episode schizophrenia patients: a double-blind placebocontrolled pilot study. Eur Neuropsychopharmacol 2004;14:332–336. 84 Ko YH, Joe SH, Jung IK, Kim SH: Topiramate as an adjuvant treatment with atypical antipsychotics in schizophrenic patients experiencing weight gain. Clin Neuropharmacol 2005;28:169–175. 85 Kim JH, Yim SJ, Nam JH: A 12-week, randomized, open-label, parallel-group trial of topiramate in limiting weight gain during olanzapine treatment in patients with schizophrenia. Schizoph Res 2006;82:115–117. 86 Nickel MK, Nickel C, Muehlbacher M, Leiberich PK, Kaplan P, Lahmann C, Tritt K, Krawczyk J, Kettler C, Egger C, Rother WK, Loew TH: Influence of topiramate on olanzapine-related adiposity in women: a random, double-blind, placebo-controlled study. J Clin Psychopharmacol 2005;25:211–217.
45
87 Egger C, Muehlbacher M, Schatz M, Nickel M: Influence of topiramate on olanzapine-related weight gain in women: an 18-month follow-up observation. J Clin Psychopharmacol 2007;27:475–478. 88 Wu RR, Zhao JP, Guo XF, He YQ, Fang MS, Guo WB, Chen JD, Li LH: Metformin addition attenuates olanzapine-induced weight gain in drug-naive first-episode schizophrenia patients: a double-blind, placebo-controlled study. Am J Psychiatry 2008; 165:352–358. 89 Wu RR, Zhao JP, Jin H, Shao P, Fang MS, Guo XF, He YQ, Liu YJ, Chen JD, Li LH: Lifestyle intervention and metformin for treatment of antipsychoticinduced weight gain: a randomized controlled trial. JAMA 2008;299:185–193. 90 Klein DJ, Cottingham EM, Sorter M, Barton BA, Morrison JA: A randomized, double-blind, placebocontrolled trial of metformin treatment of weight gain associated with initiation of atypical antipsychotic therapy in children and adolescents. Am J Psychiatry 2006;163:2072–2079. 91 Baptista T, Martínez J, Lacruz A, Rangel N, Beaulieu S, Serrano A, Arapé Y, Martinez M, de Mendoza S, Teneud L, Hernández L: Metformin for prevention of weight gain and insulin resistance with olanzapine: a double-blind placebo-controlled trial. Can J Psychiatry 2006;51:192–196. 92 Baptista T, Rangel N, Fernández V, Carrizo E, El Fakih Y, Uzcátegui E, Galeazzi T, Gutiérrez MA, Servigna M, Dávila A, Uzcátegui M, Serrano A, Connell L, Beaulieu S, de Baptista EA: Metformin as an adjunctive treatment to control body weight and metabolic dysfunction during olanzapine administration: a multicentric, double-blind, placebo-controlled trial. Schizophr Res 2007;93:99–108. 93 Borovicka MC, Fuller MA, Konicki PE, White JC, Steele VM, Jaskiw GE: Phenylpropanolamine appears not to promote weight loss in patients with schizophrenia who have gained weight during clozapine treatment. J Clin Psychiatry 2002;63:345–348. 94 Poyurovsky M, Pashinian A, Gil-Ad I, Maayan R, Schneidman M, Fuchs C, Weizman A: Olanzapineinduced weight gain in patients with first-episode schizophrenia: a double-blind, placebo-controlled study of fluoxetine addition. Am J Psychiatry 2002;159:1058–1060.
95 Bustillo JR, Lauriello J, Parker K, Hammond R, Rowland L, Bogenschutz M, Keith S: Treatment of weight gain with fluoxetine in olanzapine-treated schizophrenic outpatients. Neuropsychopharmacology 2003;28:527–529. 96 Lu ML, Lane HY, Lin SK, Chen KP, Chang WH: Adjunctive fluvoxamine inhibits clozapine-related weight gain and metabolic disturbances. J Clin Psychiatry 2004;65:766–771. 97 Poyurovsky M, Isaacs I, Fuchs C, Schneidman M, Faragian S, Weizman R, Weizman A: Attenuation of olanzapine-induced weight gain with reboxetine in patients with schizophrenia: a double-blind, placebocontrolled study. Am J Psychiatry 2003;160:297–302. 98 Poyurovsky M, Fuchs C, Pashinian A, Levi A, Faragian S, Maayan R, Gil-Ad I: Attenuating effect of reboxetine on appetite and weight gain in olanzapine-treated schizophrenia patients: a doubleblind placebo-controlled study. Psychopharmacology (Berl) 2007;192:441–448. 99 Goodall E, Oxtoby C, Richards R, Watkinson G, Brown D, Silverstone T: A clinical trial of the efficacy and acceptability of D-fenfluramine in the treatment of neuroleptic-induced obesity. Br J Psychiatry 1988;153:208–213. 100 Modell W, Hussar AE: Failure of dextroamphetamine sulfate to influence eating and sleeping patterns in obese schizophrenic patients: clinical and pharmacological significance. JAMA 1965;193:95–98. 101 Ding G, Yu G, Zhang J, Liang S, Liu L, Huang P, Chen H, Xiao A, Li X, Cai Y: The therapeutic effects of ling gui zhu gan tang mixture in 50 psychotic patients with obesity induced by the psychoactive drugs. J Tradit Chin Med 2005;25:25–28. 102 Duggal HS: Psychotic symptoms associated with topiramate: cognitive side effects or worsening of psychosis? J Clin Psychiatry 2004;65:1145. 103 Hamoui N, Kingsbury S, Anthone GJ, Crookes PF: Surgical treatment of morbid obesity in schizophrenic patients. Obes Surg 2004;14:349–352. 104 Sarwer DB, Cohn NI, Gibbons LM, Magee L, Crerand CE, Raper SE, Rosato EF, Williams NN, Wadden TA: Psychiatric diagnoses and psychiatric treatment among bariatric surgery candidates. Obes Surg 2004;14:1148–1156. 105 Clinical guidelines on the identification, evaluation, and treatment of overweight and obesity in adults: executive summary. Expert panel on the identification, evaluation, and treatment of overweight in adults. Am J Clin Nutr 1998;68:899–917.
Leslie Citrome, MD, MPH Nathan S. Kline Institute for Psychiatric Research 140 Old Orangeburg Road Orangeburg, NY 10962 (USA) Tel. +1 845 398 5595, Fax +1 845 398 5483, E-Mail
[email protected] 46
Citrome · Vreeland
Thakore J, Leonard BE (eds): Metabolic Effects of Psychotropic Drugs. Mod Trends Pharmacopsychiatry. Basel, Karger, 2009, vol 26, pp 47–65
Glucose Abnormalities in Schizophrenia, Bipolar and Major Depressive Disorders Chris Bushe Eli Lilly and Company, Basingstoke, UK
Abstract Glucose abnormalities have been recognised to be important parameters that may predict many adverse cardiovascular outcomes in the general population. Their description is complex and more so in patients suffering from mental disorders where the more formal measurements of fasting data become more difficult to obtain routinely. Despite this, there is clarity that abnormal glucose parameters may be found in at least 30% patients with severe forms of mental illness. This chapter aims to describe the available literature and attempt to better understand the plethora of data relating to glucose that derives from patients suffering from chronic mental disorders such as schizophrenia and mood disorders. Copyright © 2009 S. Karger AG, Basel
Glucose abnormalities have been recognised to be important parameters that may predict many adverse cardiovascular outcomes in the general population. Their description is complex and more so in patients suffering from mental disorders where the more formal measurements of fasting data become more difficult to obtain routinely. Despite this, there is clarity that abnormal glucose parameters may be found in at least 30% patients with severe forms of mental illness [1]. This chapter aims to describe the available literature and attempt to better understand the plethora of data relating to glucose that derives from patients suffering from chronic mental disorders such as schizophrenia and mood disorders. Where glucose abnormalities (diabetes and pre-diabetes) fit into the metabolic syndrome and indeed the validity of the metabolic syndrome, using current definitions in predicting adverse outcomes does not fall into the remit of this chapter nor do other forms of psychotic disorders.
Diabetes and Pre-Diabetes in the General Population
Diabetes (DM) type 2 is highly prevalent worldwide in 2008 and will have increasing prevalence through until at least 2025 coincident with a global obesity epidemic [2].
Diabetes type 1 is a very different disorder that is predominantly genetically inherited and in which obesity is not an important risk factor. Worldwide there are substantial regional variations with Africa having the lowest prevalence. In Europe, prevalence will increase from around 6% in 2008 to 8% in 2025 in the 20–79 age group and in some regions will increase to over 10% [2]. The worldwide prevalence of DM was 5.1% in 2003 and will rise to 6.3% by 2005. In the UK it is estimated that by 2010 DM will be diagnosed in 7% of men and 5% of women [3]. In addition, impaired glucose tolerance (IGT) will rise from 8.2% in 2003 to 9% in 2005. The sheer relevance of these data is encapsulated in the single statistic that in 2007 3.8 million deaths will be due to DM globally [4]. Complications of DM are of concern on many levels in that peripheral vascular disease (amputation risk is increased by 15–40 times compared with the general population), retinopathy and cardiovascular disease are mostly also irreversible once initiated, although there are some modern management techniques that benefit patients [4]. Many subjects with glucose abnormalities remain undiagnosed which may increase these figures further and within this group there may be very specific populations, including the family members of patients with severe mental illness who could be targeting for screening. Using data from NHANES, the prevalence of DM and impaired fasting glycaemia (IFG) in adults >20 years of age was 5.9% and 6.1%, respectively, in 2000 (but DM increasing to 15% in >60 years), which increased to 8.3% when adding undiagnosed DM [5].
Prevalence of Glucose Abnormalities in Schizophrenia
Diabetes It has been established for over 100 years that glucose abnormalities are more frequent in mentally ill subjects [6] and without doubt predate the emergence of antipsychotic treatments. The precise prevalence and incidence rates are only starting to be described and are in part dependent on the cohort evaluated and the year of study. Screening for glucose abnormalities has increased since 2000 and data prior to then are less complicated in their interpretation. A lifetime DM prevalence of 15% was reported from the Schizophrenia Patient Outcomes Research Team (PORT) in the USA in data collected dirung the mid-1990s prior to atypicals [7]. The data were stringently collected with subjects being paid for their time spent in interview. In 2002, a prevalence rate of 18% in an outpatient cohort was noteworthy for the clear findings that the prevalence increased with age, as in the general population, with the highest prevalence of >25% in the 60- to 69-year cohort [8]. In some cohort studies, prevalence rates can be compared with those in the general population at that time. Prevalence rates of 15.8% in Italy, in a schizophrenia cohort admitted to a longterm care facility, vastly exceeded the then current rates of 2–3% in the Italian general population [9]. There can be little doubt that not all patients were screened and
48
Bushe
screening bias needs to be factored in. For example, younger subjects may be more likely to be admitted and hence receive routine glucose tests. Prevalence in certain ethic groups may also be variable. Subramamiam et al. [10] reported on an inpatient cohort in Singapore where chart review found the DM prevalence rate to be 4.9% in a large cohort receiving typicals. Following an oral glucose tolerance test (OGTT), however, the overall prevalence rose to 21% emphasising the value of OGTT as a diagnostic tool. In the 50- to 59-year-old cohort, the prevalence was 50%. Screening bias, however, needs to be factored into any naturalistic data set. Data from the UK Maudsley and other local London trusts derived in 2002–2003 found that only 41% of inpatients had some form of glucose testing on case notes review [11, 12]. The actual prevalence rate for glycaemic abnormalities was more accurately determined as 15.6% (39 of 250 tested subjects) as opposed to a case notes review prevalence of 6.4% (39 of 606 subjects) of whom the vast majority had DM (n = 37) and only 2 patients had IFG. There were no reported cases of IGT strongly implying that this test had not been applied frequently. The very low prevalence of any form of pre-diabetic abnormality might suggest that abnormal glucose levels outside of diagnostic diabetic levels had not been considered relevant and not recorded. At that time, there may have been a lower awareness of the prognostic value and morbidity of pre-diabetes. Data from USA in a chronic inpatient cohort of over 13,000 subjects with schizophrenia finds a prevalence of 11% [13]. These prevalence rates need to be contextualised with what is happening worldwide as, in the USA, the age-adjusted prevalence of obesity (BMI >30) in adults increased from 22.9% in 1988–1994 to 30.5% in 1999– 2000 [14], and the age-adjusted prevalence of diagnosed diabetes in adults increased from 5.1% in 1997 to 6.5% in 2002 [5]. A lot of prevalence data are complex to interpret. A recent cross-sectional study of 210 subjects, despite reporting a prevalence for DM of only 4.8%, excluded subjects undergoing treatment for DM and the latter numbers are not often reported. When the additional 5.2% with IFG (>6.1 mmol/l) are included, the interpretation of these data might be that 10% had ‘hidden’ glucose abnormalities [15]. To calculate the true DM prevalence, it would be necessary to also know how many excluded subjects had current diagnoses of DM or pre-diabetes. Over the last 5 years, data have begun to emerge on the incidence of DM in schizophrenia cohorts. These data are also complex to interpret as the incidence and prevalence of DM in the general population is rising. The question to focus on is whether the incidence rate of DM is rising faster in subjects with serious mental illness (SMI) than in the general population. This is not a question that can be answered at the present as screening bias in both populations remains both a caveat and a confounder. Some evidence that the incidence rate of DM is rising to a greater extent in the SMI population than in the general population derives from a large USA cohort study of inpatients with schizophrenia and bipolar disorder. In this cohort, the prevalence of diabetes increased from 6.9% in 1997 to 14.5% in 2004 [16] with associated increases in incidence rates of diabetes from 0.9% in 1997 to 1.8% in 2004. During the same period,
Glucose Abnormalities in Chronic Mental Disorders
49
the incidence of diabetes rose from 0.51 to 0.72% in the USA general population [17], and the age-adjusted prevalence of diabetes in adults increased from 5.1% in 1997 to 6.5% in 2002 [5]. Traditionally, in schizophrenia the lowest DM rates are measured in the age group 18–44 years [8]. In 2003, in the USA cohort the DM prevalence of 10.0% contrasts with 2.3% for the corresponding age cohort in the general population [16]. In a naturalistic Belgian cohort (n = 183), a 3-month incident rate of 4.4% (n = 8) for DM was reported using OGTT [18]; however, when screening the original cohort (n = 220), 5.5% had previously been diagnosed with DM. In the DM incident cohort 5 of the 8 subjects had forms of pre-diabetes at the study outset. These data are not dissimilar to other reported incidence rates. The same group have reported an annual incidence rate of 4.2% in an identically screened stable schizophrenia population [19], and other annual incidence rates of 3.2–3.6% [20] and 4.4% [21] in subjects newly commenced on antipsychotics have also been reported. Incidence rates are higher (4.7 and 6.9%) and more difficult to interpret from diagnostically mixed samples [22, 23]. In a large USA inpatient sample of chronic subjects, the annual incident rates during 2000–2002 were lower, i.e. 1.25–1.5%. This may reflect better previous screening [13]. The Belgian group under De Hert has produced some interesting naturalistic data from a consecutive prospective cohort of 415 schizophrenia subjects [24]. They report that the difference in DM prevalence between schizophrenia subjects and a control population increases with increasing age being 1.6% in the 15- to 25-year cohort and 19.2% in the 55- to 65-year cohort. Using OGTT, they identified more DM cases than by fasting glycaemia (FG) alone which would have identified only 46.2% of DM cases. In comparing cohorts, they divided subjects into cohorts 5.6 mmol/l (the inclusion criteria for metabolic syndrome IDF) was found to increase from 8 to 40.5%.
Pre-Diabetes Although usage of this term may be technically inaccurate (as diabetes is not an inevitable outcome), it is useful to capture under a single metric the numbers of patients who cannot be diagnosed as DM but who have abnormal glucose values to some extent. Fasting glucose cannot always be obtained pragmatically in SMI patients both for psychiatric and logistical reasons. Many patients are unable to successfully fast perhaps due to failure to understand the precise and exact nature of the fast needed. In the UK, this has been acknowledged in two consensus group reports in 2004 and 2005 [25, 26]. In addition, although the ADA clearly state that an OGTT is an important diagnostic tool to diagnose all forms of glucose abnormality they also recognise the impracticality of general use. As a consequence, many current glucose measurements undertaken outside of a formal randomised control trial (RCT) are non-fasting and the sensitivity/specificity of a single glucose test may differ little between fasting
50
Bushe
and non-fasting [27]. Non-fasting also confers the benefit of no risk of overestimation of an abnormality. Pre-diabetes thus captures abnormal non-fasting levels, IGT and IFT as well as cases where repeat measurements are needed to confirm DM. Data are relatively recent on the prevalence and incidence of pre-diabetes. The highest reported prevalence is 30–31% IGT [10, 28]. These data derive from a prospective cross-sectional studies and revealed large amounts of undiagnosed hyperglycaemia as almost all these patients were previously considered to be euglycaemic. The Sernyak study provided further insight into difficulties of obtaining fasting glucose samples. In a total of 647 outpatients receiving antipsychotics, an attempt was made to obtain a fasting sample. However, only 23.6% of the samples were fasting and 76.4% were random. Despite excluding all known diabetics 30.1% of the fasting samples had abnormally elevated glucose (24.8% fasting plasma glucose (FPG) 5.6–6.9 mmol/l; 5.2% (n = 8) FPG >7.0 mmol/l). In contrast, case-note reviews have reported prevalence rates as low as almost zero [11], implying that there may have been a failure to document abnormal glucose levels failing to reach a diabetogenic threshold. Psychiatrists in the UK may have been unaware of the prognostic importance of a pre-diabetic diagnosis. In one of a series of naturalistic data sets (n = 183) from Belgium [18] using OGTT, pre-diabetic abnormalities were reported in 16%. Following 3 months of antipsychotic treatments pre-diabetic abnormalities had regressed in 40% of the cases (n = 12) and had developed newly in 29 cases, giving a pre-diabetes prevalence at 3 months of 26%, emphasising the value of screening using OGTT where feasible (although usually impractical in SMI patients) as opposed to fasting samples. Treatment-naïve cohorts have also suggested an elevated rate of glucose abnormalities compared to controls with Ryan et al. [29] reporting a 15% prevalence of IFG with no DM cases.
Mood Disorder
There is a general perception that glucose abnormalities in bipolar disorder are not dissimilar to schizophrenia. The recent NICE guidelines for bipolar disorder emphasise the importance of glucose monitoring [30]. The problem is untangling bipolar disorders (all types) from treatment disorders and unipolar depression. Accordingly, definitive data on glucose metabolism is less defined with a complete absence of treatment-naïve data.
Bipolar Disorder and Glucose Although there are fewer data than in schizophrenia there is an acceptance that glucose abnormalities are more frequent in bipolar subjects than in the general population and there is likely to be a similar prevalence to the schizophrenia population
Glucose Abnormalities in Chronic Mental Disorders
51
[9, 31]. A chart review of inpatients reported 26% prevalence [32]. Recent data are also supportive that metabolic syndrome of which glucose often forms an important parameter is also more prevalent than in the general population with rates of 30–32% reported [33, 34].
Depression and Glucose There has been awareness for many years that some connection exists between depression and diabetes. The increased usage of antipsychotics in treating major depression may complicate the analysis of the relationship between the disease and the onset of diabetes. A recent large USA chart review found that in a cohort of psychiatric inpatients receiving antipsychotics, the largest diagnostic group was major depressive disorder (27.6% of the sample) comparing with schizophrenia (13.8%) with 54.2% of these depressed subjects receiving antipsychotics [35]. Further data are needed to confirm if this finding might apply in all countries. An additional complexity is the diagnostic uncertainty between unipolar depression and various forms of bipolar disorder. Pre-diabetes is often found in subjects with various depressive diagnoses considered to be euglycaemic when tested [28]; however, the first observation that depression is associated with diabetes was made in 1684 [36]. A recent review concluded that the point prevalence of depression in diabetes is 11% and that dependent on methodology prevalence may be as high as 28.5% [37, 38]. Meta-analysis found from 20 studies that odds of depression in a diabetic cohort were twice that of the non-diabetic cohort (OR = 2.0, 95% CI 1.8–2.2) [38] and no differences in depression prevalence between type 1 and type 2 DM could be determined. Importantly, depression is also associated with worsening of glycaemic control and remission with a reduction in glycosylated haemoglobin [39]. Cortisol was originally suggested as a putative pathophysiological mechanism; however, hyperglycaemia itself may worsen the depressive illness [40]. The precise relationship between depression, its treatment and glycaemia is complex to fathom out but when cohorts with major depressive disorder are followed the incidence of diabetes is higher than anticipated [37]. The reasons are unclear but may involve increased activity of the sympathoadrenal system, antidepressant medications, inactivity or other physical health changes mediated by the depressive illness. An algorithm has been proposed for management utilising essentially the common treatments but emphasising the importance of the comorbidity of these bidirectional illnesses [37].
Potential Pathophysiological Mechanisms for Glucose Elevation
Unravelling the relative qualitative and quantitative components to determine precise causation has proven complex and thus far remains unknown. From the general
52
Bushe
population, we know that type 2 diabetes may take up to 10 years from initial abnormality to elevated glucose. No clinical trial in modern-day psychiatry exceeds 3 years [41]. Thus, proposed mechanisms are speculative and include weight-gain associated increases in insulin resistance, a direct toxic drug effect on muscle or adipose tissue leading to insulin resistance, stimulation of hepatic glucose production and inhibition of insulin release through pancreatic toxicity. The quantitative role of abnormally elevated amounts of visceral fat [42, 43] present in treatment-naïve subjects (over three times greater than that in controls) and any other additional genetic predisposition to DM versus any treatment or lifestyle associated association is unknown. Some recent data have led some to hypothesize that in SMI patients there may be a maximal threshold beyond which obesity may not impact greatly on insulin resistance [44]. Data from a 6-month randomised trial of schizophrenia subjects with baseline BMI 28–30 randomised to olanzapine or risperidone found no worsening or differences between the groups on parameters such as insulin sensitivity and disposition index (measure of pancreatic beta-cell functioning) using intravenous glucose tolerance testing. Treatment-emergent weight gain and small increases in visceral fat were observed in both treatment groups The hypothesis suggested was that these data were consistent with a non-linear relationship between obesity and insulin resistance. There have been a number of other studies, albeit small cohorts that have evaluated a variety of antipsychotics in forms of testing such as hyperinsulinaemic euglycaemic clamps [45], comparison of visceral fat [42, 43], and intravenous glucose tolerance tests [46, 47]. Some studies were carried out in healthy subjects, some in schizophrenia subjects and importantly some were limited to non-obese subjects. The studies, however, have been cross-sectional and open to confounding. In studies in healthy subjects [45], a significant confounder exists that the schizophrenia metabolism is clearly very different from the healthy volunteer and results may differ in schizophrenia patients and prospective studies may be too short to be definitive in light of the above caveats. Furthermore, some studies have been restricted to male subjects only [45] with current data showing that there are important sex differences in the metabolic response to antipsychotic treatments [48]. Nevertheless, these data may be significant in at least suggesting trial designs that should begin to define the pathophysiological mechanisms involved and provide some comparative antipsychotic data. Sowell et al. [49] reported in 2002 that in a 3-arm study for 15–17 days (n = 48) comparing olanzapine, risperidone and placebo using a hyperglycaemic euglycaemic clamp, there were no significant between-group differences in insulin secretion. In a small study (n = 29), Sacher et al. [45] reported that in healthy subjects after 10 days there was a higher incidence of decreased insulin sensitivity and elevated insulin levels during treatment with olanzapine compared to ziprasidone, although baseline irregularities in the olanzapine cohort need to be considered. Using a different technique, an intravenous glucose tolerance test, Henderson’s group reported data from two studies. In the first study, a cross-sectional study, subjects taking clozapine and olanzapine were found to have significant insulin
Glucose Abnormalities in Chronic Mental Disorders
53
resistance and impairment of glucose effectiveness when compared to risperidone [46], but with no significant differences in FPG or lipid parameters. The second study compared olanzapine, quetiapine and placebo [47] using the same technique and found that whereas there were significant differences compared to placebo on all parameters, there were no significant differences between patients treated with olanzapine and quetiapine on parameters of insulin sensitivity, insulin resistance, fasting insulin and glucose levels nor lipids. Glucose effectiveness was, however, lower in the olanzapine cohort (p = 0.03). There was an absence of significant change in pancreatic beta-cell function in olanzapine subjects. Finally, also using IVGT, one study reported that adiposity levels (BMI and waist circumference) were strongly related to insulin resistance [50]. Not all studies are in agreement, however. A 2-week randomised study in schizophrenia subjects randomised to olanzapine or risperidone found no differences in insulin sensitivity, insulin or glucose levels or glucose effectiveness. However, subjects taking olanzapine were found to have a decreased insulin secretory response to a hyperglycaemic challenge [51] a similar finding to a study in dogs treated with olanzapine which found impaired beta-cell compensation [52]. Other studies have shown contrasting results with no evidence that healthy subjects treated for 2 weeks with olanzapine or risperidone had any impairment of the acute insulin secretory response [53]. There are some data suggesting that subjects with schizophrenia may not be metabolically identical to control subjects with a recent study finding that insulin homeostasis was different in clozapine obese subjects compared to non-psychiatric obesity being significantly worse [54]. Currently available data do not clearly define the changes in glucose metabolism that may be observed in patients receiving longer-term treatment with antipsychotics. Ideally, future data should be derived from prospective, longer-term studies done in schizophrenia subjects and comparing commonly used atypical antipsychotics? Results of longer-term studies may differ from findings in short-term trials
Genetics and Glucose Abnormalities There is consistency that in schizophrenia subjects a positive family history of DM is found significantly more often than in a control population [9, 55, 56]. Recent data report prevalence figures of 31 and 34% [18, 57]. These data are also consistent with data from USA in which family history of diabetes in the general population has a significant powerful, independent and graded association with diabetes [58]. There are clear ethnic and geographical variations in the prevalence of diabetes worldwide and data are perhaps best considered when comparative data in that country are available. In 2006, the prevalence of type 2 DM in Barcelona was 8% [59]. In 2008, in 34 treatment-naïve subjects with non-affective forms of psychosis, a positive history of DM in either parent (27%) was found significantly more often than in controls (8%) [55].
54
Bushe
Perhaps the most powerful evidence of a genetic linkage derives from the finding that in the only study of its type a high point prevalence of IGT was found in firstdegree relatives (18.2%) and treatment-naïve schizophrenia subjects (10.5%) compared with healthy controls (0%) [60]. This study further emphasised the difficulty of carrying out studies in treatment-naïve subjects where cohort sizes are often modest and where selection of the most appropriate glucose parameter is difficult. Despite finding no significant differences between the cohorts in fasting glucose and HBA1c levels, both subjects and their relatives had elevated insulin levels and evidence of insulin resistance using HOMA-IR. There are a number of studies that suggest a genetic overlap between diabetes and schizophrenia and bipolar disorder [61]. Some preliminary data from a small study in subjects with schizophrenia support a role of the MTHFR genotype in determining a predisposition to insulin resistance [62]. Glucose levels and other parameters have tended to show abnormalities in treatment-naïve subjects; however, not all studies are consistent in this finding [29, 63–66]. A consistent finding from the Dublin group has been elevation in glucose levels and increased abdominal fat in their treatment-naïve subjects [29, 42, 43]. The underlying mechanism has, however, remained undefined. Recently, it has been proposed that deficient IGF-1 levels may underlie insulin resistance in schizophrenia [67]. In a relatively large study of 88 subjects, subjects with schizophrenia had higher insulin levels and greater insulin resistance than controls with evidence of lower IGF-1 levels. Insulin levels were elevated even after controlling for cortisol. IGF-1 has been postulated as being involved in the pathogenesis of schizophrenia [68]. Similarly, in 45 first-episode subjects either treatment-naïve (n = 15) or receiving antipsychotic treatments for mostly less than 4 weeks with a normal baseline BMI of 24.29 (SD 4.06). Although the mean FPG was significantly greater than in the control cohort (p = 0.0467), there were no differences in insulin levels although they were numerically higher in the schizophrenia patients [64]. Categorical analyses showed elevated glucose (undefined but presumably IFG) in 2.5% of the patients and 0% of the controls. CAFÉ found no glucose differences amongst a series of atypical antipsychotics over a 1-year prospective study in firstepisode subjects with schizophrenia.
Glucose Metabolism in Patients Treated with Antipsychotics The putative role that antipsychotics may play with regard to changes in glucose metabolism has been discussed in a plethora of published data. Broadly, these fall into the categories of reviews and opinions [25, 26, 69, 70], pharmaco-epidemiological studies analysing existing data retrospectively [8, 71], and prospective trials in which glucose data have been collected. What is currently absent is a single RCT in which glucose metabolism is the primary endpoint of concern other than clamp studies.
Glucose Abnormalities in Chronic Mental Disorders
55
Pharmaco-Epidemiological Studies In general terms, these have provided inconsistent findings for a number of clear reasons. Glucose abnormalities can only be detected by blood sampling and rates of screening for glucose have until recently been very low. Even in centres of excellence such as the Maudsley in London, UK, glucose screening was being undertaken in only 41% of inpatients receiving antipsychotics [11]. Furthermore, there are many confounders that should be adjusted for before attempting to compare rates of glucose changes in patients treated with antipsychotics including diagnosis, family history, weight, length of treatment, ethnicity, concomitant medications and gender, and many other potential confounders. Smoking has recently been defined as a major risk factor for type 2 diabetes [72] with a meta-analysis concluding that the risk of diabetes is increased by a relative risk (RR) of 1.61 (1.43–1.80) for heavy smokers (>20 cigarettes/day) with even former smokers having a RR of 1.23 (1.14–1.33). Rates of smoking in subjects with schizophrenia are high and cigarette consumption is also high [73–76]. Smoking is another confounder that has not been adjusted for in these studies.
Glucose Data from Prospective Clinical Trials
In 2004, the first review paper on glucose and schizophrenia data derived from prospective RCTs was published and concluded that at that time none of these data showed significant differences in changes in glucose parameters during treatment with different antipsychotics [77]. The paper further hypothesized that in some patients glucose elevations occur for reasons other than weight gain. Glucose data are collected during the majority of clinical trials and represent a routine safety parameter. The interest in glucose, insulin and glycosylated haemoglobin, however, mainly post-dates the currently marketed antipsychotics and hence did not represent a primary endpoint for any of their registration trials. Despite this, a great quantity of glucose data has been reported in many publications. In 2007, a systematic review of the then published glucose data reported that prospective glucose data were available from over 6,000 patients that participated in 22 RCTs [78]. The conclusion was that there were no consistent significant differences in changes in glucose parameters among patients treated with antipsychotics. Where any significant differences were reported, they were often in trials in which antipsychotics were used in doses 50–100% greater than the maximum licensed or labelled dose [79, 80] and trial methodologies and the reporting of the metabolic data made data interpretation complex. At present, there are now more than 30 RCTs with published glucose data and the data remain broadly similar in that no consistent significant differences between antipsychotics in individual RCTs have been reported. There are, however, a few studies that do show some differences, but these studies have unusual inclusion criteria. In an 8-week treatment-resistant study on schizophrenia where risperidone or placebo was added to clozapine treatment, the
56
Bushe
risperidone cohort had a greater increase in fasting glucose (p = 0.04) [81]. The baseline mean glucose in both cohorts was >5.6 mmol/l and thus in the range for IFG and the baseline mean PANSS was around 100. The significance of these data may also lie in the choice of parameter for analysis. Whereas there were no significant differences between the cohorts at 8 weeks in their mean glucose values the change from baseline was significant. During the study, a number of subjects (6/25 in the risperidone/clozapine cohort; 4/25 placebo/clozapine) had glucose levels >7 mmol/l at 8 weeks and were potentially diabetic. Clinicians may have found this saga not only complex but difficult to follow. The complexities and weaknesses of the retrospective data need explanation [84] and the prospective data have not always been easily accessible. One of the earliest large comparative data sets was glucose data over 6 months derived from an RCT involving olanzapine and aripiprazole. These data were salient and important as they demonstrated that although significant weight gain was observed in the olanzapine cohort (3.6 kg) and a mean weight loss was observed in the aripiprazole cohort (0.9 kg), there was no significant difference in the incidence of new-onset diabetes between the two cohorts, i.e. 4.5 and 4.7%, respectively [77]. In these subjects, the non-fasting glucose had risen from 11.1 mmol/l. The data, however, were only published on a website before inclusion in the review paper [77]. Similarly, in the 2007 systematic review of prospective data [78] from the 22 studies, 16 were peerreviewed papers, 4 posters from congresses, and 2 data from Internet sites. This paper also emphasised that FPG was reported in 16 studies and non-fasting in 7. The length of the reported studies is also important. Diabetes is recognised to be a slowly developing illness in which abnormalities in insulin resistance pre-date glucose changes by over 7 years. Only 3 studies were of at least 2 years follow-up which although long in terms of schizophrenia and psychiatry is short in glucose terms. A total of 15 studies were, however, longer than 5 months in duration. The conclusion from these prospective glucose data was that there did seem to be some consistency in that no study reported consistent glucose differences between patients treated with antipsychotics, which is in contrast to the plethora of different findings in retrospective studies [84]. Citrome et al. [85] have recently reviewed many of these retrospective studies and in addition to the inability to adjust for many confounders which makes their interpretation complex, their length of follow-up as measured by length of drug exposure at time of blood sampling is no longer than in the prospective studies. There is thus a difficult and complex task to analyze any potential role of treatment with antipsychotics in the change in glucose parameters if the traditional aetiology for diabetes is correct in schizophrenia and bipolar subjects. Some investigators may regard short-term glucose data as unhelpful to report. In a recent study that reports glucose metabolism from an open-label 12- to 16-week study [86] from subjects receiving clozapine or amisulpride glucose data was not reported. The study found insulin resistance and plasma insulin levels to be elevated in the clozapine cohort.
Glucose Abnormalities in Chronic Mental Disorders
57
What the trials have shown is that with regular glucose monitoring many new cases of diabetes and pre-diabetes will be found even in subjects currently receiving no antipsychotic medication. Aripiprazole and paliperidone are the most recent licensed antipsychotics in the UK and in their trial development placebo-controlled trials have been deemed essential by regulatory agencies. In the only study reporting on FPG levels in 2004 involving aripiprazole, over 6 months incident cases of abnormal glucose >6.1 mmol/l were measured in 5.5% subjects taking aripiprazole and 10.3% subjects on placebo [77]. Recent guidelines have thus correctly emphasised the need for regular glucose monitoring regardless of the specific antipsychotic treatment [30, 87]. Two fairly distinguished clinical trials, CAFÉ and CATIE studies [80, 82, 83], form part of the current evidence base. CAFÉ found no glucose differences between antipsychotics over a 1-year period. The CATIE study has been the landmark study in the last 10 years because of its many facets, and is a long-term independent study sponsored by the NIMH [80]. Interpretation of the glucose data from the primary publication was complex as essentially the glucose levels were admixtures of fasting and random samples in the same patient at different time points and additionally glucose was not simply adjusted for length of treatments [88]. The CATIE group subsequently published their data on metabolic syndrome parameters and prevalence using fasting data only and have recently published further purely fasting glucose data [1]. Glucose parameters are best interpreted as categorical diagnoses (diabetes, IFT, IGT) and as mean changes from baseline, when comparing different antipsychotic treatment cohorts longitudinally. At entry to CATIE many subjects randomised to antipsychotic (37–48.4%) already met the NCEP criteria for metabolic syndrome [1, 48], and the glucose parameter of >5.6 mmol/l) was met by 25–35.1%. Utilising fasting glucose data only at baseline and 3 months there were no significant differences between the antipsychotic treatment groups. Rather surprisingly, the number of subjects receiving olanzapine who met the glucose criteria decreased from 35% to 27% (n = 74). Similarly, the mean fasting glucose changes at 3 and 9 months showed no significant glucose differences between the antipsychotics. A recent 4-week RCT evaluating ziprasidone and aripiprazole also reports small increases in the median FBG of 0.11 and 0.165 mmol/l, respectively [89]. Some studies suggest that patients treated with clozapine may have the highest potential risk of glucose elevation, though few data assess the severity of illness as a potential confounder. In a 10-year follow-up, 34% (n = 96) developed diabetes though the Kaplan-Meier estimate was 43% if all patients had taken clozapine for 10 years [90]. The difficulties of adequately interpreting these data remain complex without a clinical trial designed to follow-up the glucose and metabolic parameters in the long term. Although many studies report some degree of glucose elevation during treatment with most antipsychotics there are some that report decreases. In a small open-label randomised efficacy study of around 18 weeks’ duration, significant glucose reductions were reported in all cohorts. The reason, however, is salient in that the authors report that baseline glucose levels were not fasting levels even though this was intended [91].
58
Bushe
What Format of Glucose Testing Might Be Best in 2008? The diagnosis of DM usually needs a FG appraisal and if this is feasible it should be undertaken. Recent groups reviewing this topic emphasise the pragmatic difficulty of obtaining fasting samples in SMI subjects and conclude that random samples may be feasible initially [25, 26, 28]. Certainly, the sensitivity and specificity between single FG and random glucose are not so major [27]. There are pragmatic difficulties with obtaining FPG in SMI subjects. In studies that require FPG, patients are often hospitalised [11, 29, 92]. There are clear concerns that it is difficult to accurately determine the fasting status of a patient. Despite these difficulties, investigators in Belgium report few difficulties in obtaining both FPG and undertaking OGTT. They report that without OGTT many cases of diabetes and IGT will be missed [18]. The OGTT, without doubt a good tool, is not practical in many cases despite good evidence that the sensitivity and specificity of DM diagnoses will be increased. Glycosylated haemoglobin continues to cause debate but recent data do suggest that it may be of value. The importance of re-testing in accordance with ADA and WHO definitions for the diagnosis of diabetes is clear. A number of investigators have reported that subjects may not always remain in their original glucose diagnostic category [28]. In 8 subjects considered to be euglycaemic and found to have FPG >7 mmol/l and then re-tested, in only 50% (n = 4) was a diagnosis of diabetes ratified, 2 subjects falling into the ‘pre-diabetes’ group with IFG and the remaining 2 subjects having normal FPG levels. The finding in one study that even when fasting samples are intended 75% will be non-fasting [28] further emphasises the importance of taking another look at random glucose. The IDF have recently brought out guidelines on the monitoring of glucose in diabetes and emphasise the importance of controlling post-meal hyperglycaemia [93]. A 2-hour post-meal sample 75th percentile, and two (WHO, AACE) include microalbuminuria and other features of insulin resistance. For this study, the ATP III definition of MetS was used [14]. It requires the presence of at least three of five specified criteria: FBS ≥110 mg/dl, waist circumference (WC) >102 cm for men or >88 cm for women, blood pressure (BP) ≥130 mg Hg systolic/≥85 mg Hg diastolic, triglycerides ≥150 mg dl, and high-density lipoprotein cholesterol (HDL) 110 and/or DM diagnosis and/or on DM medication, increased triglycerides and/or decreased HDL cholesterol and/or diagnosis or medication for dyslipidemia. Independent variables for these analyses were: age ≥40, female, black, Latino (comparator is white), black female, black male, Latino female, Latino male, white female (comparator is white male), substance abuse/dependence, alcohol abuse/dependence, drug abuse/dependence, opioid dependence, cocaine dependence, psychotic depression, personality disorder, borderline personality disorder, prescribed antipsychotic, typical antipsychotic, atypical antipsychotic, aripiprazole, olanzapine, quetiapine, risperidone, ziprasidone, mirtazapine, venlafaxine, TCA, topiramate, valproate, lithium, clozapine or olanzapine, quetiapine or risperidone, aripiprazole or ziprasidone, H1 blockade (i.e. receiving clozapine, olanzapine, quetiapine, mirtazapine, TCA, or typical antipsychotic), 5HT2C blockade (i.e. receiving olanzapine, mirtazapine), >1 antidepressant, and >1 antipsychotic. OR with 95% CI are reported. The H1 and HT2C variables were added because both have been implicated in weight gain [17, 21–24].
92
Goethe · Szarek · Caley
Stepwise logistic regression was also used to identify any between-group differences in patients who had all ATP III measures recorded vs. those with one or more missing criteria. Independent variables in these analyses were: age ≥40 (utilized because 40 was the median age), female, black, Latino (white as comparator), substance abuse/dependence, alcohol abuse/dependence, drug abuse/dependence, psychotic depression, antipsychotic, and atypical antipsychotic. FBS was documented for 96.6% of subjects, and for approximately three-quarters of the sample WC, triglycerides, and HDL were available (78.0, 71.6 and 71.3%, respectively). Missing data (e.g. due to invalid values, patient refusal) resulted in the creation of several subsamples. For example, all five ATP III measures were available on a subset of 1,028 patients (57.9% of the sample), and from this subset the prevalence of MetS was calculated. The prevalence of each individual measure is reported for the subset of subjects having a given criterion recorded: n = 1,386 (78.0%) for WC, n = 1,715 (96.6%) for FBS, n = 1,271 (71.6%) for triglycerides, n = 1,267 (71.3%) for HDL cholesterol. All subsamples were compared to the remainder of the sample to identify potential sample bias.
Sample The sample as a whole (n = 1,776) had 1,096 (61.7%) females and 680 males; more than half were white (63.0%, n = 1119), 25.6% (n = 455) were Latino, and 11.4% (n = 202) were black. Mean age was 38.3 years (SD 11.3). The majority of patients (91.9%, n = 1,633) had recurrent depression; 36.4% (n = 646) had psychotic features. An Axis I co-diagnosis was present in 70.2% (n = 1246), and the most common was substance abuse/dependence (57.1%, 1014); 48.3% (n = 857) had a personality disorder, and the most common were personality disorder NOS (26.3%, n = 471) and borderline (15.4%, n = 274). The subset of patients with all ATP III criteria recorded were more likely to be ≥40 years of age (OR = 1.36, CI 1.13–1.65) and to be on atypical antipsychotics (OR = 1.34, CI 1.10–1.63) but less likely to have a diagnosis of alcohol abuse/dependence (OR = 0.73, CI 0.60–0.89). Adding BMI and WC as independent variables revealed that the subset of patients who had triglycerides assessed were more likely to be ≥40 years of age (OR = 1.64, CI 1.27–2.12) and to meet the WC criterion (OR = 1.34, CI 1.04–1.63) but less likely to have a diagnosis of alcohol abuse/dependence (OR = 0.57, CI 0.44–0.74). Almost all patients in this subset (99.6%) also had HDL assessed.
Results
Pharmacotherapy The pharmacotherapy for the sample is summarized in table 1. Almost all patients received an antidepressant (98.4%, n = 1747), most commonly an SSRI (70.3%, n = 1,248). The second most commonly used antidepressant was trazodone (21.2%, n = 377), but it was almost always given at bedtime in low dosages (50–150 mg) and was never used as monotherapy. A combination of antidepressants was given to 16.9% (n = 300) of the sample. Antipsychotics were prescribed for 58.3% of patients; 66 individuals were given a typical antipsychotic, but most of this subgroup (n = 44, 66.7%) also received an atypical agent. Among the atypicals prescribed, olanzapine and clozapine (those most implicated in MetS) [25, 26] were given to only 5.3% of patients (n = 94). An anticonvulsant was prescribed for 23.6% of patients (n = 419), and the most
Metabolic Syndrome in Psychiatric Inpatients on Antidepressives
93
Table 1. Pharmacotherapy for the sample Psychotropic
n (%) receiving1
Antidepressant
1,747 (98.4)
Antipsychotic
1,035 (58.3)
Anticonvulsant
419 (23.6)
Lithium Benzodiazepine
40 ( 2.3) 608 (34.2)
1
Total >100% because many patients received >1 drug; 44% (n = 783) received 2, 26.0% (n = 462) received 3, 7.2% (n = 127) received ≥4.
commonly used were gabapentin (n = 192, 10.8%), lamotrigine (n = 85, 4.8%), and valproic acid (n = 81, 4.6%).
Prevalence of MetS Table 2 displays the proportion of patients meeting each MetS criterion and compares these values with those for the general US population in the same age range (18–59 years) [19]. The prevalence found for the full syndrome (23.4%) and for the presence of at least one criterion (75.6%) were calculated for the subset of patients for whom all metabolic measures were available (n = 1,028). Among all patients studied, 14.9% (n = 264) had MetS and for 63.6% (n = 1,128) at least one criterion was present. Compared with the NHANES sample, MDD patients were more likely to have elevated glucose (χ2 = 12.18, p < 0.001), decreased HDL cholesterol (χ2 = 41.21, p < 0.001), at least one ATP III criterion present (χ2 = 12.32, p < 0.001), and MetS (i.e., at least three ATP-III criteria) (χ2 = 8.69, p = 0.003). MDD patients were less likely than the NHANES sample to meet the hypertension criterion (χ2 = 26.69, p < 0.001), but in this study a recorded HTN diagnosis was substituted for the ATP III BP criterion used in NHANES.
Treatment and Demographic Variables Associated with MetS In the logistic regressions atypical antipsychotics were not associated with either an increased risk for MetS or the presence of one or more of the five criteria (table 3). Psychotropic exposure was relevant, however. Patients receiving venlafaxine were 65% more likely to have ≥1 criteria, and those receiving two or more antidepressants
94
Goethe · Szarek · Caley
Table 2. Prevalence of MetS criteria: study sample vs. general population Criterion
Study sample 4/05–9/07 n (%)
NHANES [19] 2003–2004 n (%)
WC (M >102 cm, F >88 cm)
671 (48.4)
1,583 (46.6)
FBS ≥110 mg/dlc
246 (14.3)
167 (10.4)
Triglycerides ≥150 mg/dl
409 (32.2)
480 (30.0)
HDL cholesterol*
465 (36.6)
901 (27.0)
Dx hypertension (M 102 cm for men or >88 cm for women, Trig = triglycerides ≥150 mg/dl, HDL cholesterol 30. Comparing glycemic control measures, 14.3% had a FBS ≥110 but 19.3% were in ‘dyscontrol’ using as the criterion FBS ≥110 and/or a documented DM diagnosis and/ or receiving a DM medication. The ATP III criteria for triglycerides and HDL were present in 32.2% and 36.6%, respectively, but 43.8% of the sample had one or both one of these measures and/or a diagnosis of dyslipidemia and/or receiving a statin.
Recognition of MetS in Clinical Practice Some findings point to the possible under recognition and/or under treatment of metabolic dysfunction. For almost all patients without a DM diagnosis, FBS was 102 cm for men or >88 cm for women, Trig = triglycerides ≥150 mg/dl, HDL cholesterol 20 ng/ml for males and >25 ng/ml for females [3]. It is now known that HPRL is the commonest disorder of the hypothalamic-pituitary axis and could be caused by physiological processes as well as pharmacological agents. Furthermore, it has been established that HPRL is associated with side effects such as galactorrhoea, gynaecomastia, hypogonadism and fertility problems, including sexual dysfunction both in men and women [4, 5]. Chronic hypogonadism has been associated
with an increased risk of osteoporosis in both sexes, and a possibly increased risk of cardiovascular disorders in young women. Additionally, there are emerging concerns about a possible association of HPRL with breast cancer and pituitary tumours [6, 7]. The focus of this chapter will be to examine in detail the impact of antipsychotic medication on HPRL and in turn its effects on the physical health of patients treated for schizophrenia.
Prolactin Physiology, Regulation and Secretion
PRL a hormone in the regulation of lactation, named by Riddle et al. [2], is today known to have over 300 separate biological activities [1] including reproduction (lactation, leuteal function, reproductive behaviour) and homeostasis (immune response, osmoregulation, angiogenesis). Human PRL consists of 199 amino acids and has similar structure, binding and functional properties as growth hormone [1]. PRL synthesis and secretion in humans is mainly from lactotrope cells in the anterior pituitary gland and is largely regulated by dopamine (inhibitory), although thyrotropin-releasing hormone (PRL release) plays a small role. To a much lesser extent, PRL can be synthesised in several other locations such as the hypothalamus, placenta and mammary gland. The links between the hypothalamus and pituitary gland are important for normal PRL secretion [8] and interference with this linkage will break the inhibitory effect of dopamine resulting in HPRL.
Common Causes of Hyperprolactinaemia
Antipsychotic-associated HPRL is believed to be the commonest cause of raised PRL. However, it is also known that some physiological, pharmacological and pathological mechanisms may also increase PRL. The main physiological causes of HPRL include pregnancy, acute stress and nipple suckling [9]. Pregnancy in schizophrenia is the most likely reason for HPRL (PRL up to 240 ng/ml) and so needs to be excluded in women presenting with clinical features of HPRL. A number of drugs other than antipsychotics affect the dopamine system, and as expected are associated with HPRL including anti-emetics, some antidepressants and some analgesics [9]. Electroconvulsive therapy has also been associated with transient PRL elevation [10]. In many centres retrospective diagnosis of possible epileptic seizure is done by measuring the post-ictal PRL levels. One of the rare but important causes of HPRL is prolactinoma which usually presents with elevated PRL levels of >100 ng/ml (2,100 IU/l) [11]. Holt [9] raises the hypothesis that dysregulation of dopamine control of PRL secretion may be an important aetiological factor in the development of prolactinomas.
Impact of Hyperprolactinaemia on the General Health of Patients
117
Other disease states associated with HPRL include primary hypothyroidism, chest wall lesions, chronic renal failure, cirrhosis of the liver, ectopic secretion of PRL, seizures and cancer metastases [12].
The Impact of Antipsychotics on Prolactin Elevation
Antipsychotic blockade of D2 receptors on the anterior pituitary lactotrope is the main mechanism by which antipsychotics increase PRL. Kapur and Seeman [13] suggested that fast dissociation from the D2 receptor makes an antipsychotic more accommodating of physiological dopamine transmission, permitting an antipsychotic effect without PRL elevation. Seeman [14] showed that the blockade of D2 receptors by typical antipsychotics is more prolonged than with atypicals and this leads to the accumulation of PRL. Kapur et al. [15] hypothesised that atypical antipsychotics with a propensity for PRL elevation would show a higher pituitary versus striatal D2 receptor occupancy and in part would reflect their ease at crossing the blood-brain barrier. Additionally, it is now recognised that some of the metabolites of antipsychotics themselves may have an effect on PRL elevation such as the 9-hydroxy metabolite of risperidone which has a predominant role in PRL elevation. The work of Green et al. [16] and Kuruvilla et al. [17] support the view that schizophrenia is not associated with raised PRL elevation in unmedicated patients. PRL elevation starts with antipsychotic treatment. Research to date shows that all antipsychotics have been associated with varying degrees of HPRL [18]; however, it is still important to note that antipsychotics show differences in terms of rates, severity and sustained levels of raised PRL [4, 18]. The quality of PRL data reported in many clinical trials is limited [19]. Few trials have PRL as a primary outcome measure, categorical levels are rarely reported, and few studies report both PRL and gonadal hormones. Most studies fail to monitor blood levels for antipsychotics and hence where a decline in PRL is reported over time, it is difficult to tease out if this is a real effect or just a reflection of non- compliance. Bushe et al. [19] identified those studies reporting categorical rates of HPRL showing a range of 42–92% in females and 18–72% in males. This confirms that HPRL is a common side effect of antipsychotic medication with females being more prone. Byerly et al. [4] suggested a clinically valuable categorisation of antipsychotics into two groups: those associated with marked and sustained PRL elevation (some typical and some atypical antipsychotic) and those second-generation antipsychotics which appear to have little effect on PRL levels. There has been a long-held view that typical antipsychotics are associated with markedly high rates of HPRL; however, an examination of the literature questions this view. Bushe and Shaw [20] found that in clinical practice the rates of HPRL in male and female patients on monotherapy with typical antipsychotics were only 33%.
118
Wildgust · Kohen
Similar rates for HPRL are shown in several other studies [21, 22] thus illustrating that many patients on typical antipsychotics do not show HPRL. The rates of HPRL reported by Bushe et al. [19] were considerably lower for typicals than for the atypicals amisulpride and risperidone. There are a number of studies which have examined the long-term impact of antipsychotics on PRL levels. Meltzer and Fang [23] reported that within 72 h men and women on phenothiazines, would show 3.2- and 3.8-fold increases in PRL levels, respectively, and these would remain elevated at 1 and 3 months during the observation period. Within 48–96 h of stopping the medication PRL levels would normalise. Chouinard et al. [24] reported that there was no evidence of growing tolerance to PRL-elevating antipsychotics in those patients who had been treated for many years with these medications. In contrast, Brown and Laughren [25] reported tolerance after 4 months of treatment with typicals. The discrepancy in these findings may be associated with issues of compliance rather than tolerance. Svestka et al. [26] reported one of the few studies with atypical antipsychotics where PRL was the primary outcome measure. Amisulpride and risperidone PRL levels increased significantly in 100% of female patients as early as the first week of treatment, whereas with quetiapine and zotepine, PRL levels were reduced as early as the first week after treatment. In the same study, olanzapine was associated with mild transient PRL elevation. In this study, PRL elevation did not correlate with age, menopausal status, efficacy of the antipsychotic, daily dose, serum lipids or blood glucose levels.
Clinical Adverse Effects of Prolactin
Halbreich and Kahn [3] proposed that many of the clinical adverse consequences of HPRL could be accounted for by its effects on the HPG axis. HPRL is associated with suppression of gonadotropin pulsatile release, which in turn inhibits the release of luteinizing hormone (LH) and follicle-stimulating hormone (FSH) from the pituitary leading to suppression of ovarian and testicular function in turn leading to hypogonadism [3, 27]. This potentially further led to a range of menstrual disorders, reduced fertility in both sexes, decreased sexual function in men and women including decreased libido, increased risk of cardiovascular disorders, galactorrhoea, anxiety and depression. Halbreich and Kahn [3] also postulated that adverse events such as an increased risk of breast cancer were possibly associated with HPRL. This hypothesis is supported by Harvey et al. [6] who identified that PRL has a direct action on breast tissue where it can act as a mitogen in the mammary gland. Furthermore, PRL stimulates the growth and motility of human breast cancer and acts as a potent survival factor protecting human breast cancer cells from apoptosis.
Impact of Hyperprolactinaemia on the General Health of Patients
119
The last decade has seen a new research focus on the secondary impact of antipsychotics on gonadal hormones and hypogonadism through HPRL in schizophrenia. Smith et al. [22] reported the impact of typical antipsychotics, with a 2-year minimum exposure on patients with schizophrenia, finding that 75% of the females and 34% of the males have HPRL. The rates of hypogonadism were 85% and 6.4% in females and males, respectively. This highlights that PRL levels are a relatively good indicator of risk of hypogonadism, but in females suggests that HPRL levels at the top end of the normal range may also indicate hypogonadism. Howes et al. [28] investigated 103 patients with schizophrenia or schizoaffective disorder with a medium treatment duration of 3.3 years with antipsychotics. The rates of hypogonadism in females was 79%, and 92% of the women had hypo-oestrogenism and 28% of the men showed hypotestosteronism.
Common Side-Effects Associated with Antipsychotic-Induced Hyperprolactinaemia
Gynaecomastia and Galactorrhoea It is important to note that there are no standardised methods for measuring galactorrhoea. Beumont et al. [29] describe massaging the breast for 3 min and then measuring the volume of milk produced. Others use a milk pump to extract milk and or just simply ask the patient. Prior to the introduction of antipsychotic medication in the 1950s neither galactorrhoea nor gynaecomastia had been noted as important complaints from patients with schizophrenia. However, Robinson [30] found an incidence rate for galactorrhoea of 10%, while Plante and Roy [31] reported an incidence rate of 57%. Tolis et al. [32] reported 65 cases of galactorrhoea and found that there was no correlation between PRL levels and the frequency and severity of galactorrhoea. Beumont et al. [29] pointed out that the correlation between raised PRL and lactation is not a direct one as other factors were involved in the initiation and maintenance of galactorrhoea. More recent studies suggest that the incidence of galactorrhoea in clinical practice is lower than earlier reports. Wesselmann and Windgassen [33] completed one of the first prospective studies of galactorrhoea in schizophrenia in 150 patients. They reported that an incidence rate of 14% and prevalence rate of 19% and galactorrhoea occurred between the 7th and 75th days after initiating treatment. Only 28% of their patients spontaneously disclosed these side effects to their psychiatrists as they found this embarrassing. Underreporting and under recognition of symptoms could be one of the underlying factors in the great variety of reported prevalence and incidence of this adverse effect. Wesselmann and Windgassen [33] stated that more than half of his female patients who experienced galactorrhoea closely linked this to their femininity. They had 1 patient who misinterpreted this as pseudo-pregnancy. Galactorrhoea is reported much less commonly in males with
120
Wildgust · Kohen
rates around 1%. However, Halbreich and Kahn [3] reported that up to 33% of men with HPRL actually had galactorrhoea on examination. It is clear that galactorrhoea should not be left to self-reporting and be included in the physical examination of the patient.
Menstrual Cycle and Fertility Menstrual dysfunction associated with psychoses was recognised well before the introduction of antipsychotics [34]. These early observations supported the view that illness and possibly life style factors also played a role in menstrual abnormalities, separate from the effects of antipsychotics on low oestrogen via hypogonadism. The rates of amenorrhoea vary widely between studies, ranging from very low figures to as high 78% as reported by Smith et al. [22], and averaging around the 30% level. This wide variation in rates may be a consequence of the definition of amenorrhoea. It has been defined as the temporary or permanent absence of menstruation for more than 6 months. Wong and Seeman [35] defined amenorrhoea as 3 consecutive missed periods. Hence, many pharmacological studies in schizophrenia designed for 12 weeks or less would miss clinical signs of adverse events of the menstrual cycle. Wong and Seeman [35] highlighted that 90% of the women linked normal menstruation with good health and subjects with irregular menses had significantly lower self-esteem than those with normal menses (p < 0.01). In the same study, patients with amenorrhoea had higher PRL levels than those without (57 vs. 27 ng/ml). Smith et al. [22] followed up patients who had been stabilised on typical antipsychotics for at least 2 years measuring both PRL levels as well as gonadal hormones. Although the average dose of antipsychotics in chlorpromazine equivalents was only 384 mg, 75% of the females had PRL levels greater than the upper limit of normal, 36% had amenorrhoea, 32% oligomenorrhoea and 36% had apparent normal menstruation; however, only 37% of these women were actually ovulating. Furthermore, this study showed that HPRL in women was correlated with the degree of suppression of the HPG axis. Howes et al. [28] found similarly high levels of intermittent anovulatory cycles (92%) in a population of premenopausal females with a median exposure to antipsychotic of 3.3 years. In clinical practice, menstrual disturbance is the most obvious feature of HPRL and has been traditionally used as a clinical sign for raised PRL. Smith et al. [22] and Howes et al. [28] demonstrated that many women with apparently regular menstruation would not be ovulating, indicating a hypogonadal state. With the appearance of antipsychotics with a lower propensity for raised PRL levels and amenorrhoea, clinicians should also recognise that switching to these medications may initiate ovulation and put the patient at risk of unplanned pregnancy.
Impact of Hyperprolactinaemia on the General Health of Patients
121
Decreased Bone Mineral Density and Osteoporosis Osteoporosis is defined by the WHO as at least a fall of 2.5 SDs below the mean peak bone density for young adults and osteopenia is defined as bone density between 1.0 SD. Prior to the introduction of antipsychotics, there were not many reports of fractures or osteopenia/osteoporosis in patients with schizophrenia, except for a few case reports of hip fractures in patients undergoing unmodified electroconvulsive therapy. A great deal of our current knowledge and understanding about the risks of developing osteopenia/osteoporosis or fractures associated with HPRL comes from animal studies and reports of patients with pituitary tumours. Little is known about the putative role of PRL on bone cells and bone formation in humans [36]. Adler et al. [37] investigated the effects of PRL excess and estrogen deficiency on bone in rats and concluded that HPRL with osteoporosis is likely to be due to PRL-induced hypogonadism rather than a direct effect of PRL on calcium homeostasis. Amenorrhoea associated with HPRL was the first recognised model of functional hypogonadal osteoporosis [38] showing an association with decrease cortical and trabecular bone density. Other recognised associations are smoking, lack of exercise and lithium-induced hyperparathyroidism [39]. Klibanski et al. [38] were the first to highlight that young women were vulnerable to developing osteopenia/osteoporosis. There are several studies of patients diagnosed with pituitary tumours who report increased rates of bone loss. Biller et al. [40] found that bone density decreased significantly in HPRL women who were amenorrheic for more than 20 months. This suggests that studies in treatment pathways in schizophrenia should continue for at least 2 years to pick up these phenomena. Further, it does confirm the clinical importance of amenorrhoea as a risk factor for osteoporosis and osteopenia. Greenspan et al. [41] confirmed that osteoporosis developed in men with hyperprolactinaemic hypogonadism. O’Keane [27] points out that prior to 2000, there were no studies in schizophrenia which examined the relationship between hypogonadism, secondary to HPRL and bone density. The clinical trials in this area have been well described and reviewed by several authors including Leucht et al. [42] and Byerly et al. [4]. Overall, there is a consistency of findings amongst these reviewers. Albeit limited by small cohorts and short duration, these studies show that patients with schizophrenia, on antipsychotics over a prolonged period of time, report increased rates of osteoporosis and osteopenia. Kishimoto et al. [43] reports a study in 74 male patients (mean age 58.9) with schizophrenia, investigating the possible causes of reduced bone mineral density. They report that 37% had osteopenia and 27% osteoporosis, 87% of the subjects had HPRL and vitamin D levels were normal. Exercise and vitamin D did not protect these patients from lower bone mineral density. The high PRL group showed low
122
Wildgust · Kohen
levels of FSH and LH, and significantly low gonadal hormones. The results of this study support the hypothesis that HPRL impacts on the HPG axis which contributes to bone loss and the greatest bone loss correlated with the increased duration of HPRL. Vekemans and Robyn [44] highlighted that in men PRL levels rise with age, whereas in women PRL levels decline with age and by the time men are 60, their normal PRL levels are higher than women of similar ages. This observation raises the important question of increased risks in older men. Howard et al. [45] completed the first large- scale population study to investigate patients with a history of schizophrenia and risk of hip fracture. The method used was a case-controlled study comparing hip fractures in the general practice research database (n = 16,341) with matched controls (n = 29,889). The key finding was that hip fractures were significantly associated with a diagnosis of schizophrenia (OR 1.73; 95% CI 1.32–2.28) and independently associated with PRL elevating antipsychotics (OR = 2.6; 95% CI 2.43–2.78). This is one of the main studies to show that schizophrenia has to be recognized as a significant risk factor for hip fractures.
Breast Cancer Leucht et al. [42], in a systematic review of physical illness in schizophrenia, found that population studies which examined the relationship between schizophrenia and breast cancer were inconclusive. They also examined the question of whether high PRL levels promote breast cancer. Their findings supported the view that the evidence for this was discordant. This chapter explores new findings which were not included in the review by Leucht et al. [42]. Catts et al. [46] completed one of the first meta-analysis of cancer incidence rates in schizophrenia which included 100,000 patients and 70,000 parents and 73,000 siblings. This meta-analysis showed that the incidence of breast cancer was significantly increased in female patients SIR = 1.12 (CI 1.02–1.23; p = 0.02). In parents of patients with schizophrenia or their siblings, the pooled data for all cancers were shown to be significantly reduced in parents: SIR = 0.90 (CI 0.88–0.93) and siblings SIR = 0.89 (CI 0.84–0.94). These findings are consistent with genes associated with schizophrenia, being protective against cancers. Hippisley-Cox et al. [47] report an epidemiological study using the QRESEARCH database (UK primary care clinical records). The population consisted of roughly 4 million patients with nearly 18.7 million person year’s observation. The study showed an increased risk of breast cancer in patients with schizophrenia (adjusted OR 1.52 for deprivation, smoking, obesity and use of other medications, 95% CI 1.10–2.11). This study differs from many of the earlier studies as it included many patients aged >50 years. Since breast cancer is more common in older women, it is likely that a greater powered study would be required to detect increased rates of breast cancer in younger women.
Impact of Hyperprolactinaemia on the General Health of Patients
123
Hankinson et al. [48] investigated the risk of breast cancer associated with high PRL by doing a series of epidemiological studies now well known as the US Nurse Health Study. This study cohort was establishes in 1976 with 121,700 US female registered nurses (30–55 years) and followed up every 2 years. From 1989 through 1990 blood samples for different blood biochemistry including PRL results were collected from 32.826 women. Hankinson et al. [48] observed a significant association with observed PRL levels and postmenopausal breast cancer (highest versus lowest quartile multivariate relative risk of 2.02; 95% CI 1.24–3.31). Hankinson along with Tworoger have now published a series of papers which show increase risk of breast cancer both in premenopausal and postmenopausal women. This is probably the strongest clinical evidence supporting the view that elevated PRL is a risk factor for breast cancer. Harvey et al. [6] believe that the balance of evidence from in vitro and animal studies now supports the view that raised PRL is associated with breast cancer. Furthermore, they challenge the long-held view that findings from animal data may not be applicable to humans. These findings are now being included in some of the summary of product characteristics in the US for some of the antipsychotics. There are some confounds with the PRL story, as there is laboratory evidence that trifluoroperazine inhibits the development of cancer cells. Halbreich et al. [49] identified high rates of breast cancer in patients with chronic schizophrenia who were screened with mammography. Wernicke et al. [50] completed one of the first studies to assess breast screening for patients with schizophrenia. Overall, they found psychiatric patients with a diagnosis of psychosis (OR = 0.33, CI 0.18–0.61; p < 0.01) were less like to attend for breast screening than the general population. Both studies highlight the importance of breast screening in patients with schizophrenia. In conclusion, findings from a meta-analysis suggests that schizophrenia maybe protective of breast cancer in parents and siblings, but breast cancer rates were significantly increased in female patients. Epidemiological studies by Hankinson and Tworoger provide strong evidence supporting the thesis that higher levels of PRL potentially put patients at increased risk of breast cancer.
Hyperprolactinaemia and Sexual Dysfunction Sexual dysfunction is associated with a wide variety of possible mechanisms. Gitlin [51] described the possible effects of a direct CNS effect on the neurotransmitter system, sedation secondary to histamine, a peripheral effect resulting in priaprism, and hormonal effects primarily through HPRL. In patients with schizophrenia, it is extremely difficult to disentangle the sexual dysfunction caused by HPRL from the psychopathology of schizophrenia and the impact of antipsychotics and other medication on a variety of receptor systems such
124
Wildgust · Kohen
as serotonin and histamine. Weizman et al. [52] conducted a study to look at sexual dysfunction associated with HPRL patients with renal failure undergoing haemodialysis. They noted that patients with high levels of PRL (130 ng/ml) as compared to patients with low levels of PRL (36.3 ng/ml) were clinically impotent. On treatment with bromocriptine, 4 males and 1 female showed a restoration of sexual desire and potency. These findings support the hypothesis that HPRL can be a major cause for reversible sexual dysfunction. Smith et al. [53] published one of the first controlled studies investigating the frequency and underlying mechanisms of sexual dysfunction in people on antipsychotics. Sexual dysfunction occurred in 45% of patients taking antipsychotics and 17% of non-treated controls. They concluded that HPRL was the main cause of sexual dysfunction in females (with 75% of the women being HPRL) whereas in males (with 34% of the men being HPRL) autonomic side effects were a dominant cause of sexual dysfunction but in those men with HPRL this superseded other causes of sexual dysfunction. Howes et al. [28] investigated the relationship between sexual function and gonadal hormones. Although this study showed high rates of sexual dysfunction and high rates of hypogonadism (92% women and 28% men) there was no correlation between sexual function and gonadal hormones which indicated that this is not the main aetiological factor in these cases. Costa et al. [54] reported no differences in sexual function between patients treated with the atypical antipsychotic olanzapine and conventional antipsychotics but they did show differences in the rate of normalisation of hormone levels. This illustrates that sexual function is a complex multi-factorial activity controlled by many different neurotransmitters and not just PRL.
Clinical Management of Hyperprolactinaemia
Historically, it was considered that a raised PRL in association with typical antipsychotics was almost inevitable. This gave rise to a nihilistic perspective, which ignored HPRL in patients unless they complained of adverse events. With the advent of newer antipsychotics with a lower risk of HPRL this perspective is no longer valid. It is now recognised that raised PRL is not benign and the risk of HPRL can be reduced through screening and planned antipsychotic treatments. HPRL and its clinical side effects have not received all the attention they deserve. The symptoms and sexual side effects of HPRL have not been fully clinically recognized or sufficiently investigated. There are still male and female patients who are not checked for their PRL levels, who do not complain to their GPs or the community key workers in the mental health teams and who suffer silently. The first step in recognition of the symptoms and side effects in the community will be the training of GPs and non-medical professionals in primary care trusts in gathering the information and recognizing the side effects in patients who may be unwilling to discuss the issues or who may not be aware of the impact of the medication.
Impact of Hyperprolactinaemia on the General Health of Patients
125
Community mental health nurses, key workers and community pharmacist should be involved in assessing the problem. The next step should be to measure PRL levels routinely, on initiating antipsychotic treatment and during the stabilisation period as part of patients physical health screen. This should be followed by training in recognition and follow-up of the longer-term systemic and metabolic side effects of the medication including establishment of osteoporosis and breast problems as early as possible. For all these the patients, their families and carers should be involved in understanding the side effects and in asking the relevant questions. Medical management and psychiatrists should make the case for financial resources when necessary. There are several guidelines on the monitoring and management of PRL in schizophrenia, but few offer practical advice [55]. Peveler et al. [56] put together some clinical recommendations on antipsychotic-associated HPRL based on a critical appraisal of the current literature. Their main recommendation was that PRL elevation appeared to be of greatest concern in people under the age of 25 years as they are at greatest risk of subsequent osteoporosis. HPRL should be avoided in those with a history of breast cancer, possibly prostate cancer, prolactinomas and those diagnosed with osteoporosis. HPRL should also be avoided in women planning pregnancy. Clinicians should consider giving patients information about the risks of elevated PRL, particularly osteoporosis, reduced fertility, menstrual irregularities and sexual dysfunction. Peveler et al. [56] suggest that in any patients presenting with PRL levels greater than 150 ng/ml, a prolactinoma should be considered and the patients should be referred to a specialist endocrinologist. Hopefully, the data of PRL will now been translated from basic sciences to clinical practice and will lead to better quality of life and life expectancy for patients at risk of developing HPRL.
References 1
2
3
4
126
Freeman ME, Kanyicska B, Lerant A, Nagy G: Prolactin: structure, function, and regulation of secretion. Physiol Rev 2000;80:1523–1631. Riddle O, Bates RW, Dykshorn SW: The preparation, identification and assay of prolactin: a hormone of the anterior pituitary. Am J Physiol 1933; 105:191–216. Halbreich U, Kahn LS: Hyperprolactinemia and schizophrenia: mechanisms and clinical aspects. J Psychiatr Pract 2003;9:344–353. Byerly M, Suppes T, Tran Q, Baker RA: Clinical implications of antipsychotic-induced hyperprolactinaemia in Patients with schizophrenia and bipolar spectrum disorder. J Clin Psychiatry 2007;27:639– 661.
5
6
7
8 9
Haddad PM, Wieck A: Antipsychotic-induced hyperprolactinaemia mechanisms, clinical features and management. Drugs 2004;64:2291–2314. Harvey PW, Everett DJ, Springall CJ: Adverse effects of prolactin in rodents and humans: breast and prostate cancer. J Psychopharmacol 2008;22:20–27. Szarfman A, Tonning JM, Levine JG, Doraiswamy PM: Atypical antipsychotics and pituitary tumours: a pharmacovigilance study. Pharmacotherapy 2006; 26:748–758. Ben-Jonathan N, Hnasko R: Dopamine as a prolactin (PRL) inhibitor. Endocr Rev 2001;22:724–763. Holt RIG: Medical causes and consequences of hyperprolactinaemia. J Psychopharmacol 2008;22: 28–37.
Wildgust · Kohen
10 Pritchard PB 3rd, Wannamaker BB, Sagel J, Nair R, Devillier C: Endocrine function following complex partial seizures. Ann Neurol 1983;14:27–32. 11 Serri O, Chik CL, Ur E, Ezzat S: Diagnosis and management of hyperprolactinemia. CMAJ 2003;169: 575–581. 12 Liu JK, Couldwell WT: Contemporary management of prolactinomas. Neurosurg Focus 2004;16:1–11. 13 Kapur S, Seeman P: Does fast dissociation from the D2 recptor explain the action of atypical antipsychotics? A New Hypothesis. Am J Psychiatry 2001; 158:360–369. 14 Seeman P: Atypical antipsychotics: mechanism of action. Focus, 2004. Am Psychiatr Assoc 2004;2: 48–58. 15 Kapur S, Langlois X, Vinken P, Megens AAHP, De Coster R, Andrews JS: The differential effects of atypical antipsychotics on prolactin elevation are explained by their differential blood-brain disposition: a pharmacological analysis in rats. J Pharmacol Exp Ther 2002;302:1129–1134. 16 Green AI, Faraone SV, Brown WA: Prolactin shifts after neuroleptic withdrawal. Psychiatr Res 1990;32: 213–219. 17 Kuruvilla A, Peedicayil J, Srikrishna G, Kuruvilla K, Kanagasabapathy AS: A study of serum prolactin levels in schizophrenia: comparison of males and females. Clin Exp Pharmacol Physiol 1992;19:603– 606. 18 Turrone P, Kapur S, Seeman MV, Flint AJ: Elevation of prolactin levels by atypical antipsychotics. Am J Psychiatry 2002;159:133–135. 19 Bushe C, Shaw M, Peveler R: A review of the association between antipsychotics and hyperprolactinaemia. J Psychopharmacol 2008;22:46–55. 20 Bushe C, Shaw M: Prevalence of hyperprolactinaemia in a naturalistic cohort of schizophrenia and bipolar outpatients during treatment with typical and atypical antipsychotics. J Psychopharmacol 2007;21:768–773. 21 Kinon BJ, Gilmore JA, Liu H, Halbreich UM:. Prevalence of hyperprolactinemia in schizophrenic patients treated with conventional antipsychotic medications or risperidone. Psychoneuroendocrinology 2003;28(suppl 2):55–68. 22 Smith S, Wheeler MJ, Murray R, O’Keane V: The effects of antipsychotic-induced hyperprolactinaemia on hypothalamic-pituitary-gonadal-axis. J Clin Psychopharmacol 2002;22:109–114. 23 Meltzer HY, Fang VS: The effect of neuroleptics on serum prolactin in schizophrenic patients. Arch Gen Psychiatry 1976;33:279–286. 24 Chouinard G, Annable L, Jones BD, Collu R: Lack of tolerance to long-term neuroleptic treatment in dopamine tuberoinfundibular system. Acta Psychiatr Scand 1981;64:353–362.
25 Brown WA, Laughren TP: Tolerance to the prolactin-elevating effect of neuroleptics. Psychiatry Res 1981;5:317–322. 26 Svestka J, Synek O, Tomanová J, Rodáková I, Cejpková A: Differences in the effect of secondgeneration antipsychotics on prolactinaemia: six weeks open-label trial in female in-patients. Neuro Endocrinol Lett 2007;28:881–888. 27 O’Keane V: Antipsychotic-induced hyperprolactinaemia, hypogonadism and osteoporosis in the treatment of schizophrenia. J Psychopharmacol 2008;22:70–75. 28 Howes OD, Wheeler MJ, Pilowsky LS, Landa S, Murray RM, Smith S: Sexual function and gonadal hormones in patients taking antipsychotic treatment for schizophrenia or schizoaffective disorder. J Clin Psychiatry 2007;3:361–367. 29 Beumont PJ, Gelder MG, Friesen HG, Harris G, Mackinnon P, Mandelbrote BM, Wiles JM: The effects of phenothiazines on endocrine function: I. Patients with inappropriate lactation and amenorrhoea. Br J Psychiatry 1974;124:413–419. 30 Robinson B: Breast changes in the male and female with chlorpromazine or reserpine therapy. Med J Aust 1957;44:239–241. 31 Plante N, Roy P: Galactorrhea and neuroleptics. Laval Med 1967;38:103–107. 32 Tolis G, Somma M, Van Campenhout J, Friesen H: Prolactin secretion in sixty-five patients with galactorrhea. Am J Obstet Gynecol 1974;118:91–101. 33 Wesselmann U, Windgassen K: Galactorrhea: subjective response by schizophrenic patients. Acta Psychiatr Scand 1995;91:152–155. 34 Kohen D, Wildgust HJ: Evolution of hyperprolactinaemia as an entity. J Psychopharmacol 2008;22: 6–11. 35 Wong J, Seeman MV: Prolactin, menstrual irregularities, quality of life. Schizophr Res 2007;91:270– 271. 36 Clément-Lacroix P, Ormandy C, Lepescheux L, Ammann P, Damotte D, Goffin V, Bouchard B, Amling M, Gaillard-Kelly M, Binart N, Baron N, Kelly PA: Osteoblasts are a new target for prolactin: analysis of bone formation in prolactin receptor knockout mice. Endocrinology 1999;140:96–105. 37 Adler RA, Evani R, Mansouri A, Krieg RJ Jr: Relative effects of prolactin excess and estrogen deficiency on bone in rats. Metabolism 1998;47:425–428. 38 Klibanski A, Neer RM, Beitins IZ, Ridgeway EC, Zervas NT, McArthur JW: Decreased bone density in hyperprolactinemic women. N Engl J Med 1980; 303:1511–1514. 39 Misra M, Papakostas GI, Kliabanski A: Effects of psychiatric disorders and psychotropic medication on prolactin and bone metabolism. J Clin Psychiatry 2004;65:1607–1618.
Impact of Hyperprolactinaemia on the General Health of Patients
127
40 Biller BM, Baum HB, Rosenthal DI, Saxe VC, Charpie PM, Klibanski A: Progressive trabecular osteopenia in women with hyperprolactinemic amenorrhea. J Clin Endocrinol Metab 1992;75:692– 697. 41 Greenspan SL, Neer RM, Ridgway EC, Klibanski A: Osteoporosis in men with hyperprolactinemic hypogonadism. Ann Intern Med 1986;104:777–782. 42 Leucht S, Burkard T, Henderson JH, Maj M, Sartorius N: Physical Illness and Schizophrenia. London, Cambridge Press, 2007. 43 Kishimoto T, Watanabe K, Shimado N, Makita K, Yagi G, Kashima H: Antipsychotic-induced hyperprolactinemia inhibits the hypothalamo-pituitarygonadal axis and reduces bone mineral density in males patients with schizophrenia. J Clin Psychiatry 2008;69:385–391. 44 Vekemans M, Robyn C: Influence of age on serum prolactin levels in women and men. Br Med J 1975;iv:738–739. 45 Howard L, Kirkwood G, Leese M: Risk of hip fracture in patients with a history of schizophrenia. Br J Psychiatry 2007;190:129–134. 46 Catts VS, Catts SV, O’Toole BI, Frost AD: Cancer incidence in patients with schizophrenia and their first degree relatives: meta-analysis. Acta Psychiatr Scand 2008;117:323–336. 47 Hippisley-Cox J, Vinogradova Y, Coupland C, Parker C: Risk of malignancy in patients with schizophrenia or bipolar disorder. Arch Psychiatry 2007;60:1368–1376. 48 Hankinson SE, Willett WC, Michaud DS, Manson JE, Coldit GA, Longcope C, Rosne B, Speize FE: Plasma prolactin levels and subsequent risk of breast cancer in postmenopausal women. J Natl Cancer Inst 1999;91:629–634.
49 Halbreich U, Shen J, Panaro V: Are chronic psychiatric patients at increased risk for developing breast cancer? Am J Psychiatry 1996;153:559–560. 50 Werneke U, Horn O, Maryon-Davis A, Wessely S, Donnan S, McPherson K: Uptake of screening for breast cancer in patients with mental health problems. J Epidemiol Community Health 2006;60:600– 605. 51 Gitlin MJ: Psychotropic medications and their effects on sexual function: diagnosis, biology and treatment approaches. J Clin Psychiatry 1994;55: 406–413. 52 Weizman R, Weizman A, Levi J, Gura V, Zevin D, Maoz B, Wijsenbeek H, Ben David M: Sexual dysfunction associated with hyperprolactinemia in males and females undergoing hemodialysis. Psychosom Med 1983;45:259–269. 53 Smith S, O’Keane V, Murray R: Sexual dysfunction in patients taking conventional antipsychotic medication. Br J Psychiatry 2002;181:49–55. 54 Costa AMN, Silvia de Lima M, Filjp SR, Reis de Olivera I, Jesus Mari J: A naturalistic, 9 month followup, comparing olanzapine and conventional antipsychotics in sexual function and hormonal profile of males with schizophrenia. J Psychopharmacol 2006; 20:6–19. 55 Citrome L: Current guidelines and their recommendations for prolactin monitoring in psychosis. J Psychopharmacol 2008;22:90–97. 56 Peveler RC, Branford D, Citrome L, Fitzgerald P, Harvey PW, Holt RIG, Howard L, Jones I, Kohen D, O’Keane V, Pariante C, Pendlebury J, Smith S, Yeomans D: Antipsychotic associated hyperprolactinaemia:clinicalrecommendations.JPsychopharmacol 2008;22:98–103.
Hiram J. Wildgust, PhD Hiram Consulting 11, Cricketers Close Ackworth WF7 7PW (UK) Tel./Fax +44 1977 615 208, E-Mail
[email protected] 128
Wildgust · Kohen
Author Index
Afzal, A. 82
Leonard, B.E. 1
Bushe, C. 47
Morenkova, A. 12
Caley, C.F. 90 Citrome, L. 25
Peveler, R.C. 66 Rasgon, N. 12
Dinan, T.G. 105 Fitzgerald, P. 105
Stemmle, P. 12 Szarek, B.L. 90
Goethe, J.W. 90
Thakore, J. 82
Holt, R.I.G. 66
Vemuri, M. 12 Vreeland, B. 25
Jiang, B. 12 Wildgust, H.J. 116 Kohen, D. 116
129
Subject Index
Amantadine, obesity management 38 Amisulpride hyperprolactinemia induction 119 obesity studies 31 Arginine vasopressin (AVP), schizophrenia levels 83, 84 Aripiprazole, obesity studies 29 Atypical antipsychotics, see also specific drugs bipolar disorder in women and insulin resistance effects 18, 19 hyperprolactinemia induction 111, 112, 119 obesity association 26, 29–31 Bariatric surgery 41 Bipolar disorder (BD) insulin resistance in women drug effects atypical antipsychotics 18, 19 carbamazepine 19 lamotrigine 19 lithium 17, 18 topiramate 19, 20 valproate 16, 17 zonisamide 19, 20 metabolic syndrome prevalence and pathophysiology 13, 14 obesity rates 14, 15 prospects for study 20 sex differences 13 metabolic syndrome association 12, 51, 52 polycystic ovarian disease association 15 Body mass index (BMI), classifications 26 Bone mineral density (BMD), hyperprolactinemia effects 122, 123 Breast cancer, hyperprolactinemia 123, 124
130
Carbamazepine bipolar disorder in women and insulin resistance effects 19 metabolic syndrome association 100 Cardiovascular disease (CVD) economic impact 68, 69 epidemiology 69 mortality 66, 68 prevention 71, 72, 76, 77 risk factors 70 screening in serious mental illness 67, 73–75, 77–80 Celecoxib, schizophrenia treatment with risperidone 6 Clozapine cytokine response 5 metabolic syndrome association 99 obesity studies 29 Cortisol, see Hypothalamic-pituitary-adrenal axis Cyclooxygenase-2 (COX-2), schizophrenia activity 6 Depression insulin resistance 52 metabolic syndrome association epidemiology 90, 91 inpatient study definitions of metabolic syndrome 91, 82, 97 limitations 101 pharmacotherapy 93, 94 prevalence 94, 98, 99 study design 91–93 treatment and demographic variables 94–96, 99, 100
metabolic syndrome recognition in clinical practice 97, 98 Dextroamphetamine, obesity management 40 Diabetes type 2 epidemiology 47, 48, 68 glycemic control 76 lifestyle modification intervention 71 risk factors 69, 70 schizophrenia association epidemiology 48–50, 60 inflammation 6, 7 pre-diabetes 50, 51 screening in serious mental illness 67, 72–75, 77–80 Diet obesity and life expectancies 27, 28 obesity management in mental illness 33 Dopamine, regulation of prolactin 108–110 Famotidine, obesity management 38 Fenfluramine, obesity management 40 Fluoxetine, obesity management 40 Gabapentin, metabolic syndrome association 100 Galactorrhea, hyperprolactinemia 120, 121 Gynecomastia, hyperprolactinemia 120, 121 Haloperidol, cytokine response 5 Hyperglycemia, see Insulin resistance Hyperprolactinemia, see Prolactin Hypothalamic-pituitary-adrenal (HPA) axis hypercortisolemia central and physical effects 85, 86 normal physiology 83 schizophrenia dysfunction 83–85 stress, obesity, and metabolic syndrome 86, 87, 100, 101 Inflammation, schizophrenia diabetes type 2 and obesity role 6, 7 leptin role 7, 8 Insulin-like growth factor-1 (IGF-1), schizophrenia and deficiency 55 Insulin resistance (IR) bipolar disorder in women drug effects atypical antipsychotics 18, 19 carbamazepine 19 lamotrigine 19 lithium 17, 18
Subject Index
topiramate 19, 20 valproate 16, 17 zonisamide 19, 20 metabolic syndrome prevalence and pathophysiology 13, 14 obesity rates 14, 15 prospects for study 20 sex differences 13 depression 52 glucose data from prospective clinical trials 56–58 glucose testing in mental illness 59, 72–75 pathophysiological mechanisms for glucose elevation antipsychotic 53, 54 genetics 54, 55 pharmaco-epidemiological studies 56 wellness program effects 59, 60 Interferon-␥ (IFN-␥), schizophrenia levels 4 Interleukin-1 (IL-1), schizophrenia levels 4 Interleukin-2 (IL-2), schizophrenia levels 4 Interleukin-6 (IL-6), schizophrenia levels 3, 4 Lamotrigine bipolar disorder in women and insulin resistance effects 19 metabolic syndrome association 100 Leptin, inflammation and obesity role in schizophrenia 7, 8 Lithium bipolar disorder in women and insulin resistance effects 17, 18 metabolic syndrome association 100 Menstrual dysfunction, hyperprolactinemia 121 Metabolic syndrome, see also Insulin resistance; Obesity bipolar disorder association 51, 52 women 13, 14 depression association epidemiology 90, 91 inpatient study definitions of metabolic syndrome 91, 82, 97 limitations 101 pharmacotherapy 93, 94 prevalence 94, 98, 99 study design 91–93
131
Metabolic syndrome (continued) treatment and demographic variables 94–96, 99, 100 metabolic syndrome recognition in clinical practice 97, 98 stress, obesity, and metabolic syndrome 86, 87, 100, 101 Metformin, obesity management 38, 39 Nizatidine, obesity management 38 Obesity atypical antipsychotic association 26, 29–31 bariatric surgery 41 bipolar disorder in women 14, 15 definitions 26, 27 dietary factors and life expectancies 27, 28 epidemiology 27 health risks 25, 26 inflammation and schizophrenia diabetes type 2 and obesity role 6, 7 leptin role 7, 8 lifestyle modification therapy in mental illness 31–36 pharmacological interventions adjunctive pharmacological interventions 37–40 orlistat 36, 37 rimonabant 37 sibutramine 37 stress, obesity, and metabolic syndrome 86, 87 Olanzapine hyperprolactinemia induction 119 metabolic syndrome association 99 obesity studies 29–31 pathophysiological mechanisms for glucose elevation 53, 54 Orlistat, obesity management 36, 37 Osteoporosis, hyperprolactinemia 122, 123 Polycystic ovarian disease (PCOD), bipolar disorder association 15 Prolactin dopamine regulation 108–110 hyperprolactinemia antipsychotic medication induction atypical antipsychotics 111, 112, 119 overview 105, 106, 118, 119 prospects for study 112, 113 typical antipsychotics 110, 111
132
clinical adverse effects breast cancer 123, 124 galactorrhea 120, 121 gynecomastia 120, 121 menstrual dysfunction 121 osteoporosis 122, 123 overview 119, 120 sexual dysfunction 124, 125 definition 116 etiology 117, 118 health risks 105, 106, 116, 117 management 125, 126 receptors and signaling 108 secretion regulation 106 structure 107 synthesis and secretion 107, 117 Quetiapine, obesity studies 31 Reboxetine, obesity management 39, 40 Rimonabant, obesity management 37 Risperidone hyperprolactinemia induction 119 obesity studies 30, 31 pathophysiological mechanisms for glucose elevation 53, 54 schizophrenia treatment with celecoxib 6 Schizophrenia diabetes type 2 association epidemiology 48–50 pre-diabetes 50, 51 glucose data from prospective clinical trials 56–58 hypothalamic-pituitary-adrenal axis dysfunction 83–85 immune changes antipsychotic drug effects 5, 6 cellular immunity 3–5 human leukocyte antigen susceptibility locus 2, 3 inflammation diabetes type 2 and obesity role 6, 7 leptin role 7, 8 insulin-like growth factor-1 deficiency 55 life expectancy 1 metabolic changes and disease 1, 2 obesity and lifestyle modification therapy 33–35 Sexual dysfunction, hyperprolactinemia 124, 125
Subject Index
Sibutramine, obesity management 37 Solutions for wellness program 33, 35 Stress, see Hypothalamic-pituitary-adrenal axis T helper cell, schizophrenia antipsychotic drug effects 5, 6 cytokine profile 4 Topiramate bipolar disorder in women and insulin resistance effects 19, 20 obesity management 39 Transforming growth factor- (TGF-), schizophrenia levels 5
Subject Index
Valproate bipolar disorder in women and insulin resistance effects 16, 17 metabolic syndrome association 99 Ziprasidone, obesity studies 29, 30 Zonisamide, bipolar disorder in women and insulin resistance effects 19, 20
133